EP3388064B1 - 2,4-thiazolidinedione derivatives in the treatment of central nervous system disorders - Google Patents

2,4-thiazolidinedione derivatives in the treatment of central nervous system disorders Download PDF

Info

Publication number
EP3388064B1
EP3388064B1 EP18173541.6A EP18173541A EP3388064B1 EP 3388064 B1 EP3388064 B1 EP 3388064B1 EP 18173541 A EP18173541 A EP 18173541A EP 3388064 B1 EP3388064 B1 EP 3388064B1
Authority
EP
European Patent Office
Prior art keywords
compounds
compound
mixture
ethoxy
thiazolidine
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP18173541.6A
Other languages
German (de)
French (fr)
Other versions
EP3388064A1 (en
Inventor
Ana María GARCÍA COLLAZO
David John Augustus ECKLAND
Maria Pilar Pizcueta Lalanza
Marc Martinell Pedemonte
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Minoryx Therapeutics SL
Original Assignee
Minoryx Therapeutics SL
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Minoryx Therapeutics SL filed Critical Minoryx Therapeutics SL
Priority to SI201531591T priority Critical patent/SI3388064T1/en
Priority to PL18173541T priority patent/PL3388064T3/en
Publication of EP3388064A1 publication Critical patent/EP3388064A1/en
Application granted granted Critical
Publication of EP3388064B1 publication Critical patent/EP3388064B1/en
Priority to HRP20210518TT priority patent/HRP20210518T1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • This invention relates to novel uses of 2,4-thiazolidinedione derivatives as medicaments in particular for the treatment of central nervous system disorders.
  • NS disorders are diseases of any component of the brain and the spinal cord.
  • NS disorders include disorders in which the nervous system is affected during the entire progression of the diseases such as neurodegenerative diseases (e.g., Alzheimer's disease, Huntington's chorea, Parkinson's disease, amyotrophic lateral sclerosis (ALS), degenerative ataxias such as Friedrich's ataxia, multiple sclerosis, multiple system atrophy and leukodystrophies), cerebrovascular diseases (e.g., global or local ischemia, intracerebral haemorrhage, stroke), seizures and epilepsy, viral diseases (e.g., meningitis, encephalitis), brain tumors and neuroinflammatory diseases.
  • NS disorders also include disorders in which the nervous system is only affected during the latest stages of the development of the disorder. These disorders comprise rare metabolic diseases such as organic acidemias or fatty acid disorders and genetic mitochondrial disorders.
  • Neurodegenerative diseases are characterised by the progressive loss of structure or function of neurons, including death of neurons. These conditions are progressive and often fatal. The process of neurodegeneration is not well understood and the diseases that stem from it have, as yet, no cures in spite of treatments being constantly sought.
  • Some neurodegenerative diseases also include an inflammatory component, such as multiple sclerosis which traditionally was considered as inflammatory mediated demyelinating diseases but, in fact, is a neurodegenerative disease in which axonal damage, neuronal death and atrophy of the central NS are the principal causes of irreversible neurological disability in patients.
  • multiple sclerosis can be considered as a neurodegenerative disease but also as a neuroinflammatory disease or autoimmune disease.
  • Leukodystrophies are a group of generic NS disorders whose main feature is the degeneration of the white matter in the brain.
  • One disorder of this group is adrenoleukodystrophy (X-linked adrenoleukodystrophy or X-ALD). This is a rare, inherited disorder that leads to progressive damage to the brain and other tissues and eventually death. This disease can be considered both as neurodegenerative and neuroinflammatory.
  • X-ALD presents three main phenotypes: (i) an adult adrenomyeloneuropathy (AMN) with axonopathy in spinal cords, (ii) cerebral adrenomyeloneuropathy with brain demyelination (cAMN), and (iii) childhood variant (cALD) characterized by severe cerebral demyelination.
  • APN adult adrenomyeloneuropathy
  • cAMN cerebral adrenomyeloneuropathy with brain demyelination
  • cALD childhood variant characterized by severe cerebral demyelination.
  • X-ALD is the most frequently inherited leukodystrophy, with a minimum incidence of 1 in 17,000 including hemizygous males and carrier females.
  • Cerebrovascular diseases are a group of brain dysfunctions related to disease of the blood vessels supplying the brain. There are four types: stroke, transient ischaemic attack (TIA), subarachnoid haemorrhage and vascular dementia.
  • TIA transient ischaemic attack
  • subarachnoid haemorrhage vascular dementia
  • Epilepsy is an unpredictable, serious and potentially fatal disorder of the nervous system. About 50 million people worldwide have epilepsy.
  • Brain tumours are generated by an abnormal and uncontrolled cell division not only in the brain (neurons or glial cells) but also in blood vessels, cranial nerves, meninges, skull, and pituitary or pineal glands. Brain tumours also include those that have spread from primary cancer cells located in other organs (metastasis).
  • Nervous system viral diseases are caused by viral infections in the NS. These infections can induce neurological dysfunction and potentially serious inflammatory diseases such as encephalitis, an inflammation of the brain itself, meningitis that results in inflammation of the meninges or myelitis that means spinal cord inflammation. Rabies, measles, mumps, poliomyelitis, herpes simplex or varicella-zoster are types of nervous system viral infections.
  • Rare metabolic diseases also known as Inborn Errors of Metabolism
  • monogenic diseases where certain metabolic pathways are perturbed thus originating dysfunctions, in many cases on the central NS. They are chronically debilitating and life-threatening conditions.
  • Genetic mitochondrial diseases can be caused by mutations in either mtDNA or nDNA, that impair mitochondrial function and typically result in very severe multisystem disease from birth, including severe manifestations on the NS.
  • Pioglitazone is a drug marketed for use in the treatment of diabetes mellitus type 2.
  • Pioglitazone is a potent agonist for peroxisome proliferator-activated receptor-gamma (PPAR ⁇ ) and it has been proposed for the treatment of some neurodegenerative diseases including Alzheimer's, Parkinson's disease, ALS and Friedreich's ataxia.
  • PPAR ⁇ peroxisome proliferator-activated receptor-gamma
  • US2013/0274295 discloses the utility of Pioglitazone in the treatment of X-ALD based on pre-clinical data. Although pre-clinical models have shown promising results, clinical trials to date have failed to show clinical benefits in any of these extremely serious conditions.
  • Pioglitazone has been associated with unwanted side effects including cardiovascular effects, fluid retention, weight gain and bladder cancer. High doses of Pioglitazone are therefore undesirable as high systemic exposure would be likely to result in serious side effects.
  • Pioglitazone is a "dirty" drug which is converted to many metabolites in vivo.
  • the metabolic pathway of Pioglitazone after oral administration has been studied in several animal species and in humans and the metabolites have been described in the literature (see e.g. Sohda et al, Chem. Pharm. Bull., 1995, 43(12), 2168-2172 ) and Maeshiba et al, Arzneim.-Forsch/Drug Res, 1997, 47 (I), 29-35 ).
  • M-I to M-VI At least six metabolites have been identified, named M-I to M-VI. Amongst these metabolites, M-II, M-III and M-IV show some pharmacological activity but are less active than Pioglitazone in diabetic preclinical models.
  • central NS disorders can be treated by 5-[4-[2-(5-(1-hydroxyethyl)-2-pyridinyl)ethoxy]benzyl]-2,4-thiazolidinedione of formula (1), the M-IV metabolite of Pioglitazone. or a salt thereof.
  • NS disorders are limited to those disorders defined in the claims.
  • the invention is based at least in part on data showing the unexpected ability of the compound of formula (1) to cross the blood brain barrier.
  • the compound of the invention has one or more desirable drug properties such as good oral bioavailability, low systemic plasma clearance, and a good volume of distribution.
  • the compound of the invention is a reasonably "clean" drug, since in vivo it only converts to 5-(4-(2-(5-acetyl-2-pyridyl)ethoxy)benzyl)-2,4-thiazolidinedione (M-III metabolite of Pioglitazone) and both are excreted. Side effects due to unwanted metabolites are therefore minimised.
  • administration of compound of formula (1), or a pharmaceutically acceptable salt is useful for the treatment or prevention of the central NS disorders defined in claim 1.
  • treatment means to ameliorate or eliminate the disease or one or more symptoms associated with said disease.
  • Treatment also encompasses ameliorating or eliminating the physiological sequelae of the disease.
  • prevention refers to the reduction in the risk of acquiring or developing a given disease or disorder, or the reduction or inhibition of the recurrence or a disease or disorder.
  • the compound of formula (1) can be named 5-(4-(2-(5-(1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione and has two chiral centres. One of them is the carbon atom in the 5-position of the thiazolidine-dione ring and the other asymmetric atom is at position 1 of the hydroxyethyl group as shown by the arrows:
  • compound of formula (1) is used to designate all possible stereoisomers, including enantiomers and diastereomers, and mixtures including racemic mixtures thereof.
  • the invention provides novel compounds (2) to (5) for use in the treatment or prevention of the central nervous system disorders defined in the claims:
  • reference to compounds (1) to (5) in the present invention is intended to designate compounds (1) to (5) having hydrogen atoms which are predominantly in the form of its isotope 1 H, i.e. no more than 1 % of the total number of hydrogen atoms per mole of compound are in the form of the 2 H isotope (deuterium), still more preferably no more than 0.015 % (which is the natural abundance of deuterium) of the total number of hydrogen atoms per mole of compound are in the form of the 2 H isotope (deuterium).
  • Mixtures (c) and (d) are particularly preferred.
  • mixtures (a) to (d) mentioned above it is particularly preferred that the two compounds mentioned in each one of the mixtures are present in equimolar quantities.
  • Said mixtures may compound also minor amounts (preferably less than 10 wt. %, more preferably less than 3 wt %, still more preferably less than 1wt. % and most preferably less than 0.1 wt.% of other compounds of formula (1).
  • a compound showing high brain-to-plasma exposure is preferred.
  • a preferred compound displays potent PPAR-gamma agonist activity but compounds with less potent PPAR-gamma agonist activity are also useful.
  • Other factors including but not limited to, pharmacological activity (other than PPAR-gamma), ADME, pharmacokinetic profile, toxicity, safety, brain distribution properties, compound accumulation in tissues, compound metabolism and clearance, genotypic variation in clearance and physicochemical properties may also be considered for the selection of a preferred compound.
  • a preferred compound has low central nervous system toxicity.
  • a preferred compound has low systemic toxicity.
  • the presence or absence of PPAR alpha activity may also be considered.
  • the compound In some cases it is desirable for the compound to result in low or no accumulation in the brain. This can reduce the risk of central nervous system toxicity and/or allow rapid reversal of drug effect in the central nervous system. In other cases, high brain accumulation with limited systemic exposure may be preferred. This can result in greater central nervous system exposure to the drug and higher efficacy. It is often advantageous for the compound not to be the subject to significant genotypic variations in clearance. This results in more consistent efficacy. These activities may be determined by use of the appropriate in vitro and in vivo assays.
  • the disorder is Friedreich's ataxia.
  • a preferred compound or mixture of compounds may be selected for a particular route of delivery. Some compounds or mixture of compounds may also be preferred based on their use to treat a particular disease.
  • the compounds of the invention can be in the form of a pharmaceutically acceptable salt.
  • pharmaceutically acceptable salt refers to salts prepared from pharmaceutically acceptable inorganic and organic acids.
  • Illustrative pharmaceutically acceptable acid addition salts of the compounds of the present invention can be prepared from the following acids, including without limitation, formic, acetic, propionic, benzoic, acetic, propionic, benzoic, succinic, glycolic, gluconic, lactic, maleic, malic, tartaric, citric, nitric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, hydrochloric, hydrobromic, hydroiodic, isocitric, xinafoic, tartaric, trifluoroacetic, pamoic, propionic, anthranilic, mesylic, napadisylate, oxalacetic, oleic, stearic, salicylic, p-hydroxybenzoic, nicotinic, phenylacetic, mandelic, embonic (pamoic), methanesulfonic, phosphoric,
  • the compounds of the invention or pharmaceutically acceptable salts may be used according to the invention when the patient is also administered or in combination with one or more of another therapeutic agent selected from antiinflammatory and analgesic agents, dopamine agonists (e.g. levodopa), MAO-B inhibitors, catechol O-methyltransferase (COMT) inhibitors, anticholinergics, other antiparkinsonians (e.g. amantadine), antiNMDA receptors (e.g. memantine), cholinesterase inhibitors, ACE inhibitors, glutamate antagonist (e.g. riluzole), antioxidants, immunomodulators (e.g.
  • fingolimod anti CD52, CD25 and CD20 monoclonal antibodies, interferon- ⁇ -1a, natalizumab, laquinimod, dimethylfumarate) chemotherapeutics, enzyme replacement therapy agents, substrate reduction therapy agents, corticosteroids, antiproliferatives (e.g. methotrexate), anticonvulsant medications, anticoagulants, antihypertensives and neuroprotectives.
  • the compounds of the invention may also be used when the patient is undergoing gene therapy, bone marrow transplantation, deep brain stimulation or radiotherapy.
  • compositions for use as defined in the claims comprising compounds (2) to (5), mixtures (a) to (d) or a pharmaceutically acceptable salt represent other embodiments of the invention.
  • Any suitable route of administration can be used.
  • any of oral, intraoral, topical, epicutaneous, subcutaneous, transdermal, intramuscular, parenteral, ocular, rectal, vaginal, inhalation, buccal, sublingual and intranasal delivery routes may be suitable.
  • Oral administration may be preferred.
  • Oral forms of pharmaceutical compositions may be solid or liquid. Suitable dosage forms may be tablets, capsules, pills, granules, suspensions, emulsions, syrups or solutions.
  • the pharmaceutical composition is a solid form selected from the group consisting of tablets, capsules, pills or granules. Particularly preferred are tablets. Oral solutions or suspensions are also preferred. These are advantageous when the patient has difficulty swallowing, for example as a result of the disease or for geriatric and paediatric use. Sublingual preparations are also advantageous.
  • an “effective” amount or a “therapeutically effective amount” of a drug or pharmacologically active agent is meant a nontoxic but sufficient amount of the drug or agent to provide the desired effect.
  • the amount that is "effective” will vary from subject to subject, depending on the age and general condition of the individual, the particular active agent or agents, and the like. Thus, it is not always possible to specify an exact "effective amount”. However, an appropriate "effective” amount in any individual case may be determined by one of ordinary skill in the art using routine experimentation. Thus, he dose of the active agent will depend on the nature and degree of the condition, the age and condition of the patient, and other factors known to those skilled in the art.
  • a typical daily dosage is from 0.1 to 200 mg, preferably from 20 to 200 mg, e.g. for an adult 10-100 mg given as a single dose with no further dosing or in multiple doses, for example one to three times per day.
  • the compounds described herein may also be administered in daily doses of from 80 to 600 mg.
  • compositions may contain conventional excipients known in the art and may be prepared by conventional methods.
  • compositions may further comprise one or more therapeutic agent.
  • Combination treatments may be administered simultaneously, sequentially or separately, by the same or by different routes, or before, during and after surgical or intervention procedures.
  • the compounds for use of the invention may be prepared by any suitable method known in the art and/or by the processes described below. It will also be appreciated that functional groups, such as amino or hydroxyl groups, present in the various compounds described, and which it is desired to retain, may need to be in protected form before any reaction is initiated. In such instances, removal of the protecting group may be the final step in a particular reaction. Suitable protecting groups for such functionality will be apparent to those skilled in the art. For specific details see " Protective Groups in Organic Synthesis", Wiley Interscience, T W Greene, PGM Wuts .
  • Any mixtures of final products or intermediates obtained can be separated on the basis of the physico-chemical differences of the constituents, in known manner, into the pure final products or intermediates, for example by chromatography, distillation, fractional crystallization, or by formation of a salt if appropriate or possible under the circumstances.
  • the compounds for use according to the invention may be prepared by the following or similar processes.
  • a mixture of compounds (2) and (4) or a mixture of compounds (3) and (5) of the invention may be prepared as in Scheme 1 but using enantiomerically pure alcohols ( IIa ) and ( IIb ) as starting materials.
  • Yet another method to prepare mixtures (c) - comprising compound (2) and (4) - and (d) - comprising compounds (3) and (5) - includes the resolution of the racemic mixture VIII using the already described methods (chiral HPLC separation, enzymatic resolution, chiral resolution, etc) followed by double bond reduction in each of the enantiomers Villa and Vlllb.
  • Mixture (b) (comprising compounds of formula (4) and (5) ) and mixture (a) (comprising compounds of formula (2) and (3) ) of the invention may be prepared by asymmetric hydrogenolysis of a compound of formula VI using for example Rhodium or Iridium catalysts in the presence of chiral ligands as shown on Scheme 4. Chiral reduction of the double bond may also be performed using biocatalysts (e.g. Rhodotorula rubra and Rhodotorula glutinis).
  • biocatalysts e.g. Rhodotorula rubra and Rhodotorula glutinis
  • LLOQ lower limit of quantification
  • Pharmacokinetic parameters were calculated using the non-compartmental analysis tool of Phoenix WinNonlin.
  • FIG. 3 shows that brain concentrations for both intravenous and oral pharmacokinetic profiles were observed until 8 hr.
  • Tmax in brain is 0.50 hr with brain-to-plasma exposure (AUClast) ratio of 0.12.
  • a cellular functional assay was performed using a human recombinant cell line cotransfected with a PPRE luciferase reporter, PPAR-y, RXR- ⁇ and coactivator DRIP205.
  • Transfected cells were treated with increasing doses of compounds. Luciferase activity was detected by alphascreen technology and normalized based on ⁇ -galactosidase activity. The results are expressed as the fold induction over the control (Rosiglitazone 10 ⁇ M). Dose response curves were obtained. Results were calculated as EC 50 that is the concentration of compound that provokes 50 % control agonist response.
  • LiHMDS (1.0 M in tetrahydrofurane, 463 ml, 0.463 mol) was added drop wise to a cooled solution of methyl 6-methylnicotinate (20 g, 0.132 mol) and ethyl acetate (82 g, 0.927 mol) in dimethylformamide at -50°C; gradually raised the temperature to r.t. and stirred at the same temperature. After 1 h, the reaction mixture was cooled to 0°C; slowly diluted with 20% sulphuric acid and heated to reflux. After 4 h, the reaction mixture was cooled to r.t. and further to 0°C and basified with potassium carbonate.
  • Methanesulphonylchloride (1.19 g, 0.01 mol) was added, drop wise, to a cooled suspension of compound IV (1.7 g, 0.008 mol) and triethylamine (1.79 ml, 0.013 mol) in dichloromethane (20 ml) at 0°C and stirred at same temperature for 1 h.
  • the reaction mixture was diluted with water (50 ml) and extracted with dichloromethane (3x50 ml).
  • reaction color lightened and additional quantities of sodium borohydride (46 mg, 1.207 mmol) and CoCl 2 .6H 2 O (22 mg, 0.096 mmol) were added and stirring was continued at r.t. After 12 h, the reaction was neutralized with acetic acid (pH ⁇ 7); diluted with water (10 ml) and extracted in ethyl acetate (3x50 ml).
  • Example 5 (mixture (c) of compounds (2) and (4) ) and (mixture (c) of compounds (3) and (4) )
  • Step 1 Synthesis of compound VIII: HCl (48 ml, 2N) was added to a solution of compound VI (10 g, 0.024 mol) in methanol (200 ml) and the mixture was heated to reflux. After 4 h of reflux, the reaction mixture was cooled to r.t. and concentrated under reduced pressure to afford a yellow solid. The solid was suspended in water (70 ml) and neutralized using a saturated NaHCO 3 solution. The resulting pale yellow precipitate was collected by filtration and vacuum dried to afford compound VIII (7.5 g; 84% yield). ES-MS [M+1] + : 371.0.
  • Step 3 A solution of NaBH 4 (77 mg, 2.02 mmol) in 0.1 N NaOH (2 ml) was added slowly to a stirred solution of compound Ent-1 (VIII) (250 mg, 0.675 mmol), dimethylglyoxime (32 mg, 0.27 mmol) and CoCl 2 .6H 2 O (16 mg, 0.067 mmol) in a mixture of water (10 ml), THF (10 ml) and 1M NaOH (0.5ml) solution at 10 °C, and the reaction mixture was stirred at r.t. for 1 h.
  • Step 3 A solution of NaBH 4 (72 mg, 1.921 mmol) in 0.1 N NaOH (2 ml) was added slowly to a stirred solution of compound Ent-2 (VIII) (237 mg, 0.64 mmol), dimethylglyoxime (30 mg, 0.256 mmol) and COCl 2 .6H 2 O (15 mg, 0.064 mmol) in a mixture of water (10 ml), THF (10 ml), and 1M NaOH (0.5ml) solution at 10 °C, and the reaction mixture was stirred at r.t. for 1 h.
  • Diastereomeric mixtures D-1 and D-2 of MIV correspond to mixtures (c) and (d) described above, but the specific diastereomers present in each diastereomeric mixture have not been assigned.
  • the assays performed include cytochrome P450 inhibition with the different isoforms, microsomal and hepatocyte stability, neurotoxicity in neural cells and hERG safety assays using a patch clamp electrophysiology measurement (FDA Draft Guidance for Industry. Drug Interaction Studies - Study Design, Data Analysis, Implications for Dosing, and Labelling Recommendations 2012, The European Medicines Agency (EMA) Guideline on the Investigation of Drug Interactions Adopted in 2012, Schroeder K et al. 2003 J Biomol Screen 8 (1); 50-64 , Barter ZE et al. 2007 Curr Drug Metab 8 (1); 33-45 , LeCluyse EL and Alexandre E 2010 Methods Mol Biol 640; 57-82 ). The results indicate a safe and favourable ADME profile for the compounds of the invention.
  • Example 8 The brain plasma ratios of Pioglitazone, MIV, Mill and Mil following oral dosing of a single administration of Pioglitazone at 4.5 mg/kg in male C57BL/6 mice.
  • the brain-plasma ratio was calculated based on levels of Pioglitazone, MIV, Mill and Mllin plasma and brain quantified at C max (maximal concentration) following oral dosing of a single administration of Pioglitazone at 4.5 mg/kg in male C57BL/6 mice.
  • the percentage brain plasma ratio was 9, 13, 7 and 1 %, respectively, for Pioglitazone, MIV, Mil and Mill as shown in the Figure 4 .
  • active metabolites Mill and Mil crossed the BBB at much lower extent than Pioglitazone as it was predicted based on the physicochemical properties of the compounds (see Table1).
  • Example 9 The brain plasma ratios of Pioglitazone and MIV and following oral dosing of a single administration of either Pioglitazone or M-IV both at 4.5 mg/kg in male C57BL/6 mice.
  • the brain-plasma ratio was calculated based on the pharmacokinetics curves of plasma and brain concentration-time profiles calculated as area under the curves of Pioglitazone and following oral dosing of a single administration of either Pioglitazone or MIV both at 4.5 mg/kg in male C57BL/6 mice.
  • the percentage brain-plasma ratio was 8 % and 12% for Pioglitazone and M4 respectively as shown in the Figure 5 .
  • M-IV shows a behavior contrary to that expected. As Mil, MIV is a monohydroxylated metabolite, but instead of decreasing around 50% its BBB penetration, the BBB penetration is 50% higher.
  • Diastereomeric mixtures D-1 and D-2 of 5-(4-(2-(5-(1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione were extracted from Swiss albino mouse plasma samples using liquid-liquid extraction (LLE) method and quantified using liquid chromatography tandem mass spectrometry (LC-MS/MS) with Electro Spray Ionization (ESI) and multiple reaction monitoring (MRM). Selected plasma and brain samples were subjected for the chiral analysis using Chiral AGP column to identify the chiral inter-conversion. Achiral bioanalytical method was employed to quantify the total M-IV present in the plasma and brain samples.
  • Formulation samples were suitably diluted with 70% methanol and the instrument response was compared against known corresponding diastereomeric mixture standard using achiral LC-MS/MS method.
  • the lower limit of quantification (LLOQ) in plasma for diastereomeric mixture D-1 and D-2 of 5-(4-(2-(5-(1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione (1) is 0.99 ng/mL.
  • Pharmacokinetic parameters were calculated using the non-compartmental analysis tool of Phoenix WinNonlin.
  • Foetuses were collected and immediately placed in ice-cold Leibovitz medium. Cortex was treated with a trypsin- EDTA solution and the dissociation was stopped by addition of Dulbecco's modified Eagle's medium (DMEM) with glucose (Pan Biotech), containing DNAse I and 10% fetal calf serum (FCS). Cells were mechanically dissociated and centrifuged. The pellet was resuspended in a defined culture medium with 10 ng/ml of brain-derived neurotrophic factor (BDNF). The cells were seeded in 96-well plates precoated with poly-L-lysine and were cultured at 37°. The medium was changed every 2 days. The cortical neurons were intoxicated with glutamate after 13 days of culture.
  • DMEM Dulbecco's modified Eagle's medium
  • FCS fetal calf serum
  • BDNF and test compound were pre-incubated with primary cortical neurons for 1 hour before glutamate exposure. Glutamate was added to a final concentration of 40 ⁇ M diluted in control medium in presence of BDNF or test compound for 20 min.
  • MAO-B increase dopamine levels in the CNS affected in Parkinson's disease without increasing levels of the other neurotransmisors (epinephrine, norepinephrine or serotonine), in contrast to no selective MAO inhibitors (MAO-A and MAO B).
  • the MAO-B inhibitors can be used also to treat depressions.
  • MAO-A and MAO-B Human recombinant monoamine oxidase proteins MAO-A and MAO-B were purchased from Sigma Aldrich (Reference M7316 and M7441 respectively). In order to monitor the MAO enzymatic activities and their inhibition rate a fluorescence based assay was used.
  • the substrate for the assay, kynuramine is non-fluorescent until undergoing oxidative deamination by MAOs resulting in the fluorescent product 4-hydroxyquinoline.
  • Kynuramine is a substrate for both MAO-A and -B (non-specific substrate).
  • Clorgiline and Deprenyl Sigma Aldrich
  • Results show that 5-(4-(2-(5-(1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione inhibits MAO B with a IC 50 of 70.5 nM. In contrast, this compound did not inhibit MAO A protein.
  • Neuroinflammation can be initiated in response to a variety of infection, traumatic brain injury, toxics, or autoimmunity in the CNS.
  • the neuroinflammatory models are characterized by proliferation of astrocytes and microglia, along with neuronal loss, is a prominent feature of many diseases of the central nervous system, including Alzheimer's Disease, Multiple Sclerosis, stroke, Parkinson, traumatic brain injury, infection and ALD ( Human Molecular Genetics, 2004, Vol. 13, No. 23 2997-3006 ).
  • Chronic inflammation is the sustained activation of glial cells and recruitment of other immune cells into the brain. It is chronic inflammation that is typically associated with neurodegenerative diseases.
  • the EAE model is a neuroinflammatory model, classically used for multiple sclerosis, which resembles and includes most of the features of the severe cerebral forms of ALD, microglial activation, brain demyelination and axonal degeneration as well. Although the ethiology of the disease is different from ALD and the EAE (a model of multiple sclerosis triggered by autoreactive CD4+ lymphocytes), the EAE model is a valuable tool for studying therapies for both ALD and multiple sclerosis ( Nature 2007; 7:904-912 ).
  • EAE autoimmune encephalomyelitis
  • mice Female C57BL/6 mice (6-8 weeks old) were injected (two 100 ⁇ L subcutaneous injections into one hind limb) with 250 ⁇ g of MOG35-55 emulsified in a 100 ⁇ L phosphate-buffered solution mixed with 100 uL of complete Freund's adjuvant containing 500 ⁇ g of Mycobacterium tuberculosis (Difco, Detroit, MI).
  • the mice received an injection of pertussis toxin (400 ng in 200 ⁇ L of a phosphate-buffered solution, intraperitoneally), a second pertussis toxin injection 2 days later, and a booster injection of MOG35-55 at 7 days.
  • Clinical signs were scored as follows: 0, no signs; 1.0, limp tail/loss of righting; 2.0, ataxia with limp tail; 3.0, paralysis of single hind limb; 4.0, paralysis of both hind limbs; 4.5, moribund; and 5.0, death. For both models, scores of 5 were noted and counted on the day of death only.
  • the compound was administered twice a day (bid) started on day 5 post immunization for 15 days at three different increasing doses.
  • MIV compound (1)
  • Average daily clinical scores from the experiment are shown in FIG 9 .
  • the clinical symptoms decreases in a dose-dependent manner, the highest doses show the maximal effect.
  • Clinical symptoms were reduced by MIV, suggesting a role for PPARgamma activation in protective effects. No body weight loss and no significant hematological toxicity at highest doses associated to treatment.
  • MIV may be effective on both diseases.
  • decreasing microglia activation provides a molecular basis for explaining why allogeneic and autologous HSCT are effective at arresting the cerebral inflammation, namely by the replacement and the functional metabolic restoration of the monocyte lineage and connect cALD and AMN phenotypes with a shared pathogenic pathway ( Human Molecular Genetics, 2012, Vol. 21, No. 5 1062-1077 ).
  • these models may be have a great potential and relevance to study the role of PPAR gamma agonists in ALD.
  • Human control and X-linked adrenoleukodystrophy fibroblasts were obtained from Coriell (Candem, USA). Cells were grown in Dulbecco's modified Eagle medium containing 10% foetal bovine serum, 100 U/ml penicillin and 100 mg streptomycin, at 37 °C in humidified 95% air/5% CO 2 , to 80-90% confluence. To perform our experiments, Dulbecco's modified Eagle medium without D-glucose, pyruvate or L-glutamine was used. Cells were cultured in this medium supplemented with 1 g/l of glucose or 1 g/l of galactose and 10% foetal bovine serum for 24 hours incubated with increasing doses of MIV (3, 10 and 30 ⁇ M).
  • MTT tetrazolium salt
  • ATP levels For the determination of ATP levels, 2x10 4 cells/well were seeded in 96 well cell culture plate in complete medium. After 16-18 h cells, were lysed in 20 ⁇ l lysis buffer and 10 ⁇ l of lysate was used to measure ATP levels using ATP determination Kit (Molecular Probes, Invitrogen). 1 ⁇ l each of the remaining lysate was used for protein measurement.
  • Results show a protective effect of MIV (compound (1)) on ALD fibroblasts based on increase in cell survival (20 % at 3, ⁇ M, vs non-treated).
  • MNDs motor neuron diseases
  • PLS progressive bulbar palsy
  • SMA spinal muscular atrophy
  • PPS post-polio syndrome
  • AMN Charcot-Marie-Tooth disease
  • Rat spinal cord (SC) motor neurons were cultured as described by Martinou (Neuron. 1992 Apr;8(4):737-44 ) and Wang (PLoS Genet. 2013;9(9 )). Briefly, pregnant female rats of 14 days gestation were killed by cervical dislocation and foetuses were collected and immediately placed in ice-cold L15 Leibovitz medium. Spinal cord was treated for 20 min at 37°C with a trypsin- EDTA. The dissociation was stopped by addition of Dulbecco's modified Eagle's medium (DMEM) with glucose (Pan Biotech), containing DNAse I and 10% fetal calf serum (FCS). Cells were mechanically dissociated and they were then centrifuged.
  • DMEM Dulbecco's modified Eagle's medium
  • FCS fetal calf serum
  • the pellet was resuspended in a defined culture medium with 10 ng/ml of brain-derived neurotrophic factor (BDNF).
  • BDNF brain-derived neurotrophic factor
  • the cells were seeded in 96-well plates precoated with poly-L-lysine and were cultured at 37°. The medium was changed every 2 days.
  • BDNF or test compound was pre-incubated with primary Spinal cord (SC) motor neurons for 1 hour before glutamate exposure. Glutamate was added to a final concentration of 10 ⁇ M diluted in control medium in presence of BDNF or test compound for 20 min. After 20 min, glutamate was washed out and fresh culture medium with BDNF or test compound was added for additional 48 hours.
  • SC Spinal cord
  • the survival evaluation was done by immunostaining. 48 after the intoxication, the cell culture supernatant was taken off and the SC motor neurons were fixed by a cold solution of ethanol (95%) and acetic acid (5%) for 5 min and permeabilized. Cells were incubated for 2 hours with a monoclonal antibody anti microtubule-associated-protein 2 (MAP-2) that stains specifically cell bodies of neurons (MAP-2) allowing study of neuron survival evaluation in the culture. This antibody was revealed with Alexa Fluor 488 goat anti-mouse IgG).
  • Test compounds were pre-incubated for 1 hour before glutamate application.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Psychology (AREA)
  • Pain & Pain Management (AREA)
  • Dispersion Chemistry (AREA)
  • Physiology (AREA)
  • Nutrition Science (AREA)
  • Rheumatology (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Diabetes (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Virology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Description

    Field of the invention
  • This invention relates to novel uses of 2,4-thiazolidinedione derivatives as medicaments in particular for the treatment of central nervous system disorders.
  • Background of the invention
  • Central Nervous System (NS) disorders are diseases of any component of the brain and the spinal cord. NS disorders include disorders in which the nervous system is affected during the entire progression of the diseases such as neurodegenerative diseases (e.g., Alzheimer's disease, Huntington's chorea, Parkinson's disease, amyotrophic lateral sclerosis (ALS), degenerative ataxias such as Friedrich's ataxia, multiple sclerosis, multiple system atrophy and leukodystrophies), cerebrovascular diseases (e.g., global or local ischemia, intracerebral haemorrhage, stroke), seizures and epilepsy, viral diseases (e.g., meningitis, encephalitis), brain tumors and neuroinflammatory diseases. NS disorders also include disorders in which the nervous system is only affected during the latest stages of the development of the disorder. These disorders comprise rare metabolic diseases such as organic acidemias or fatty acid disorders and genetic mitochondrial disorders.
  • Neurodegenerative diseases are characterised by the progressive loss of structure or function of neurons, including death of neurons. These conditions are progressive and often fatal. The process of neurodegeneration is not well understood and the diseases that stem from it have, as yet, no cures in spite of treatments being constantly sought.
  • Some neurodegenerative diseases also include an inflammatory component, such as multiple sclerosis which traditionally was considered as inflammatory mediated demyelinating diseases but, in fact, is a neurodegenerative disease in which axonal damage, neuronal death and atrophy of the central NS are the principal causes of irreversible neurological disability in patients. Thus, multiple sclerosis can be considered as a neurodegenerative disease but also as a neuroinflammatory disease or autoimmune disease.
  • Leukodystrophies are a group of generic NS disorders whose main feature is the degeneration of the white matter in the brain. One disorder of this group is adrenoleukodystrophy (X-linked adrenoleukodystrophy or X-ALD). This is a rare, inherited disorder that leads to progressive damage to the brain and other tissues and eventually death. This disease can be considered both as neurodegenerative and neuroinflammatory.
  • X-ALD presents three main phenotypes: (i) an adult adrenomyeloneuropathy (AMN) with axonopathy in spinal cords, (ii) cerebral adrenomyeloneuropathy with brain demyelination (cAMN), and (iii) childhood variant (cALD) characterized by severe cerebral demyelination. X-ALD is the most frequently inherited leukodystrophy, with a minimum incidence of 1 in 17,000 including hemizygous males and carrier females.
  • Cerebrovascular diseases are a group of brain dysfunctions related to disease of the blood vessels supplying the brain. There are four types: stroke, transient ischaemic attack (TIA), subarachnoid haemorrhage and vascular dementia.
  • Epilepsy is an unpredictable, serious and potentially fatal disorder of the nervous system. About 50 million people worldwide have epilepsy.
  • Brain tumours are generated by an abnormal and uncontrolled cell division not only in the brain (neurons or glial cells) but also in blood vessels, cranial nerves, meninges, skull, and pituitary or pineal glands. Brain tumours also include those that have spread from primary cancer cells located in other organs (metastasis).
  • Nervous system viral diseases are caused by viral infections in the NS. These infections can induce neurological dysfunction and potentially serious inflammatory diseases such as encephalitis, an inflammation of the brain itself, meningitis that results in inflammation of the meninges or myelitis that means spinal cord inflammation. Rabies, measles, mumps, poliomyelitis, herpes simplex or varicella-zoster are types of nervous system viral infections.
  • Rare metabolic diseases (also known as Inborn Errors of Metabolism) are usually monogenic diseases where certain metabolic pathways are perturbed thus originating dysfunctions, in many cases on the central NS. They are chronically debilitating and life-threatening conditions.
  • Genetic mitochondrial diseases can be caused by mutations in either mtDNA or nDNA, that impair mitochondrial function and typically result in very severe multisystem disease from birth, including severe manifestations on the NS.
  • There is an urgent need for new treatments of central NS disorders.
  • A wide variety of deuterium enriched 2,4-thiazolidinediones have been described in US 2014/0275180 . This document also discloses their prophetic use in the treatment of a variety of different diseases. However, this document fails to provide any evidence in this regard or regarding the ability of these compounds to cross the blood-brain barrier (BBB).
  • Pioglitazone is a drug marketed for use in the treatment of diabetes mellitus type 2. Pioglitazone is a potent agonist for peroxisome proliferator-activated receptor-gamma (PPARγ) and it has been proposed for the treatment of some neurodegenerative diseases including Alzheimer's, Parkinson's disease, ALS and Friedreich's ataxia. US2013/0274295 discloses the utility of Pioglitazone in the treatment of X-ALD based on pre-clinical data. Although pre-clinical models have shown promising results, clinical trials to date have failed to show clinical benefits in any of these extremely serious conditions.
  • In addition, Pioglitazone has been associated with unwanted side effects including cardiovascular effects, fluid retention, weight gain and bladder cancer. High doses of Pioglitazone are therefore undesirable as high systemic exposure would be likely to result in serious side effects.
  • Pioglitazone is a "dirty" drug which is converted to many metabolites in vivo. The metabolic pathway of Pioglitazone after oral administration has been studied in several animal species and in humans and the metabolites have been described in the literature (see e.g. Sohda et al, Chem. Pharm. Bull., 1995, 43(12), 2168-2172) and Maeshiba et al, Arzneim.-Forsch/Drug Res, 1997, 47 (I), 29-35). At least six metabolites have been identified, named M-I to M-VI. Amongst these metabolites, M-II, M-III and M-IV show some pharmacological activity but are less active than Pioglitazone in diabetic preclinical models.
  • The distribution of Pioglitazone and its metabolites in various tissues after oral administration of [14C]-Pioglitazone to rats has also been studied (Maeshiba et al, Arzneim.-Forsch/Drug Res, 1997, 47 (I), 29-35). In most tissues the concentrations of Pioglitazone and metabolites M-I to M-VI were lower than that in plasma and one of the lowest concentrations of radioactivity was found in the brain where only Pioglitazone was mainly detected.
  • Summary of the invention
  • Surprisingly it has been found that central NS disorders can be treated by 5-[4-[2-(5-(1-hydroxyethyl)-2-pyridinyl)ethoxy]benzyl]-2,4-thiazolidinedione of formula (1), the M-IV metabolite of Pioglitazone.
    Figure imgb0001
    or a salt thereof.
  • In the present invention the NS disorders are limited to those disorders defined in the claims.
  • The invention is based at least in part on data showing the unexpected ability of the compound of formula (1) to cross the blood brain barrier. In addition, the compound of the invention has one or more desirable drug properties such as good oral bioavailability, low systemic plasma clearance, and a good volume of distribution. Furthermore, the compound of the invention is a reasonably "clean" drug, since in vivo it only converts to 5-(4-(2-(5-acetyl-2-pyridyl)ethoxy)benzyl)-2,4-thiazolidinedione (M-III metabolite of Pioglitazone) and both are excreted. Side effects due to unwanted metabolites are therefore minimised.
  • Thus, according to an aspect of the invention, there is provided a compound of formula (1) or pharmaceutically acceptable salts thereof or mixture of compounds of formula (1) for use in the treatment or prevention of the central NS disorders defined in claim 1.According to another aspect the present invention provides novel compounds (2) to (5):
    • (2) (R)-5-(4-(2-(5-((R)-1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione
    • (3) (R)-5-(4-(2-(5-((S)-1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione
    • (4) (S)-5-(4-(2-(5-((R)-1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione
    • (5) (S)-5-(4-(2-(5-((S)-1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione
    or a pharmaceutically acceptable salt thereof or mixtures of one or more of the compounds (2) to (5) or pharmaceutically acceptable salts thereof for use in the treatment or prevention of the central nervous system disorders defined in claim 4. Description of the figures
    • FIG 1 represents the concentration of compound of formula (1) in plasma of a C57BL/6 mouse after a single intravenous administration of 1 mg/Kg of said compound.
    • FIG 2 represents the concentrations of compound of formula (1) in plasma of a C57BL/6 mouse after a single oral administration of 4.5 mg/Kg of said compound.
    • FIG 3 represents the concentration of compound of formula (1) in brain tissue of a C57BL/6 mouse after a single oral administration of 4.5 mg/Kg of said compound (line with circle markers) and after a single intravenous administration of 1 mg/Kg of said compound (line with square markers).
    • FIG4 represents the brain plasma ratio calculated based on levels of Pioglitazone, MIV, MIII and MII in plasma and brain quantified at Cmax (maximal concentration) following oral dosing of a single administration of Pioglitazone at 4.5 mg/kg in male C57BL/6 mice.
    • FIG 5 represents the brain plasma ratio calculated based on the pharmacokinetics curves of plasma and brain concentration-time profiles calculated as area under the curves of Pioglitazone and following oral dosing of a single administration of either Pioglitazone or MIV both at 4.5 mg/kg in male C57BL/6 mice.
    • FIG 6 represents the concentrations of mixture (c) comprising compounds (2) and (4) and mixture (d) comprising compounds (3) and (5) in plasma of a C57BL/6 mouse after a single oral administration of 4.5 mg/Kg of said mixtures.
    • FIG 7 represents the effect of compound of formula (1) in primary rat cortical neurons injured by glutamate.
    • FIG 8 represents the effect of compound of formula (1) in primary culture of sensory neurons injured by Paclitaxel (Taxol).
    • FIG 9 represents the effect of compound of formula (1) in the disability score in an in vivo efficacy study in an experimental autoimmune encephalomyelitis (EAE) of multiple mouse model.
    • FIG 10 represents the effect of compound of formula (1) in primary motor neurons injured by glutamate
    Description of the preferred embodiments
  • In the present invention the terms "compound of formula (1)", "M-IV", "MIV" and "M4" indistinctively refer to 5-[4-[2-(5-(1-hydroxyethyl)-2-pyridinyl)ethoxy]benzyl]-2,4-thiazolidinedione, which has the structure depicted above.
  • In one aspect, administration of compound of formula (1), or a pharmaceutically acceptable salt is useful for the treatment or prevention of the central NS disorders defined in claim 1.
  • The term "treatment" or "to treat" in the context of this specification means to ameliorate or eliminate the disease or one or more symptoms associated with said disease. "Treatment" also encompasses ameliorating or eliminating the physiological sequelae of the disease.
  • The term "ameliorate" in the context of this invention is understood as meaning any improvement on the situation of the patient treated.
  • The term "prevention" or "to prevent" refer to the reduction in the risk of acquiring or developing a given disease or disorder, or the reduction or inhibition of the recurrence or a disease or disorder.
    Figure imgb0002
  • The compound of formula (1) can be named 5-(4-(2-(5-(1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione and has two chiral centres. One of them is the carbon atom in the 5-position of the thiazolidine-dione ring and the other asymmetric atom is at position 1 of the hydroxyethyl group as shown by the arrows:
    Figure imgb0003
  • As used herein the term "compound of formula (1)" is used to designate all possible stereoisomers, including enantiomers and diastereomers, and mixtures including racemic mixtures thereof.
  • In another aspect, the invention provides novel compounds (2) to (5) for use in the treatment or prevention of the central nervous system disorders defined in the claims:
    • (2) (R)-5-(4-(2-(5-((R)-1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione
      Figure imgb0004
    • (3) (R)-5-(4-(2-(5-((S)-1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione
      Figure imgb0005
    • (4) (S)-5-(4-(2-(5-((R)-1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione
      Figure imgb0006
    • (5) (S)-5-(4-(2-(5-((S)-1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione
      Figure imgb0007
      or pharmaceutically acceptable salts thereof.
  • Although compounds (2) to (5) have been prepared and isolated their absolute (R/S) configuration has not yet been determined and only their optical rotation has been determined.
  • Preferably, reference to compounds (1) to (5) in the present invention is intended to designate compounds (1) to (5) having hydrogen atoms which are predominantly in the form of its isotope 1H, i.e. no more than 1 % of the total number of hydrogen atoms per mole of compound are in the form of the 2H isotope (deuterium), still more preferably no more than 0.015 % (which is the natural abundance of deuterium) of the total number of hydrogen atoms per mole of compound are in the form of the 2H isotope (deuterium).
  • In a particular embodiment the following mixtures of compound (2) to (5) are preferred:
    1. (a) Mixtures comprising compounds (2) and (3), preferably being compounds (2) and (3) the only compounds of formula (1) present in the mixtures;
    2. (b) Mixtures comprising (4) and (5), preferably being compounds (4) and (5) the only compounds of formula (1) present in the mixtures;
    3. (c) Mixtures comprising (2) and (4), preferably being compounds (2) and (4) the only compounds of formula (1) present in the mixtures; and
    4. (d) Mixtures comprising (3) and (5), preferably being compounds (3) and (5) the only compounds of formula (1) present in the mixtures.
  • Mixtures (c) and (d) are particularly preferred.
  • In the mixtures (a) to (d) mentioned above, it is particularly preferred that the two compounds mentioned in each one of the mixtures are present in equimolar quantities. Said mixtures may compound also minor amounts (preferably less than 10 wt. %, more preferably less than 3 wt %, still more preferably less than 1wt. % and most preferably less than 0.1 wt.% of other compounds of formula (1).
  • For use in the treatment of central NS disorders several factors may be considered when selecting a preferred compound. A compound showing high brain-to-plasma exposure is preferred. A preferred compound displays potent PPAR-gamma agonist activity but compounds with less potent PPAR-gamma agonist activity are also useful. Other factors, including but not limited to, pharmacological activity (other than PPAR-gamma), ADME, pharmacokinetic profile, toxicity, safety, brain distribution properties, compound accumulation in tissues, compound metabolism and clearance, genotypic variation in clearance and physicochemical properties may also be considered for the selection of a preferred compound. A preferred compound has low central nervous system toxicity. A preferred compound has low systemic toxicity. The presence or absence of PPAR alpha activity may also be considered. In some cases it is desirable for the compound to result in low or no accumulation in the brain. This can reduce the risk of central nervous system toxicity and/or allow rapid reversal of drug effect in the central nervous system. In other cases, high brain accumulation with limited systemic exposure may be preferred. This can result in greater central nervous system exposure to the drug and higher efficacy. It is often advantageous for the compound not to be the subject to significant genotypic variations in clearance. This results in more consistent efficacy. These activities may be determined by use of the appropriate in vitro and in vivo assays.
  • In a particular embodiment of the aspects of the invention described above, the disorder is Friedreich's ataxia.
  • A preferred compound or mixture of compounds may be selected for a particular route of delivery. Some compounds or mixture of compounds may also be preferred based on their use to treat a particular disease.
  • The compounds of the invention can be in the form of a pharmaceutically acceptable salt. The term "pharmaceutically acceptable salt" refers to salts prepared from pharmaceutically acceptable inorganic and organic acids.
  • Illustrative pharmaceutically acceptable acid addition salts of the compounds of the present invention can be prepared from the following acids, including without limitation, formic, acetic, propionic, benzoic, acetic, propionic, benzoic, succinic, glycolic, gluconic, lactic, maleic, malic, tartaric, citric, nitric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, hydrochloric, hydrobromic, hydroiodic, isocitric, xinafoic, tartaric, trifluoroacetic, pamoic, propionic, anthranilic, mesylic, napadisylate, oxalacetic, oleic, stearic, salicylic, p-hydroxybenzoic, nicotinic, phenylacetic, mandelic, embonic (pamoic), methanesulfonic, phosphoric, phosphonic, ethanesulfonic, benzenesulfonic, pantothenic, toluenesulfonic, 2- hydroxyethanesulfonic, sulfanilic, sulfuric, salicylic, cyclohexylaminosulfonic, algenic, β- hydroxybutyric, galactaric and galacturonic acids. Exemplary pharmaceutically acceptable salts include the salts of hydrochloric acid and hydrobromic acid.
  • The utility of the compound of formula (1), including stereoisomers (2) to (5), mixtures (a) to (d) and pharmaceutically acceptable salt thereof can be demonstrated in appropriate in vitro or in vivo assays as described in the examples.
  • The compounds of the invention or pharmaceutically acceptable salts may be used according to the invention when the patient is also administered or in combination with one or more of another therapeutic agent selected from antiinflammatory and analgesic agents, dopamine agonists (e.g. levodopa), MAO-B inhibitors, catechol O-methyltransferase (COMT) inhibitors, anticholinergics, other antiparkinsonians (e.g. amantadine), antiNMDA receptors (e.g. memantine), cholinesterase inhibitors, ACE inhibitors, glutamate antagonist (e.g. riluzole), antioxidants, immunomodulators (e.g. fingolimod, anti CD52, CD25 and CD20 monoclonal antibodies, interferon-β-1a, natalizumab, laquinimod, dimethylfumarate) chemotherapeutics, enzyme replacement therapy agents, substrate reduction therapy agents, corticosteroids, antiproliferatives (e.g. methotrexate), anticonvulsant medications, anticoagulants, antihypertensives and neuroprotectives. The compounds of the invention may also be used when the patient is undergoing gene therapy, bone marrow transplantation, deep brain stimulation or radiotherapy.
  • Pharmaceutical compositions for use as defined in the claims comprising compounds (2) to (5), mixtures (a) to (d) or a pharmaceutically acceptable salt represent other embodiments of the invention. Any suitable route of administration can be used. For example, any of oral, intraoral, topical, epicutaneous, subcutaneous, transdermal, intramuscular, parenteral, ocular, rectal, vaginal, inhalation, buccal, sublingual and intranasal delivery routes may be suitable. Oral administration may be preferred. Oral forms of pharmaceutical compositions may be solid or liquid. Suitable dosage forms may be tablets, capsules, pills, granules, suspensions, emulsions, syrups or solutions. Preferably the pharmaceutical composition is a solid form selected from the group consisting of tablets, capsules, pills or granules. Particularly preferred are tablets. Oral solutions or suspensions are also preferred. These are advantageous when the patient has difficulty swallowing, for example as a result of the disease or for geriatric and paediatric use. Sublingual preparations are also advantageous.
  • By an "effective" amount or a "therapeutically effective amount" of a drug or pharmacologically active agent is meant a nontoxic but sufficient amount of the drug or agent to provide the desired effect. The amount that is "effective" will vary from subject to subject, depending on the age and general condition of the individual, the particular active agent or agents, and the like. Thus, it is not always possible to specify an exact "effective amount". However, an appropriate "effective" amount in any individual case may be determined by one of ordinary skill in the art using routine experimentation. Thus, he dose of the active agent will depend on the nature and degree of the condition, the age and condition of the patient, and other factors known to those skilled in the art. A typical daily dosage is from 0.1 to 200 mg, preferably from 20 to 200 mg, e.g. for an adult 10-100 mg given as a single dose with no further dosing or in multiple doses, for example one to three times per day. The compounds described herein may also be administered in daily doses of from 80 to 600 mg.
  • The pharmaceutical compositions may contain conventional excipients known in the art and may be prepared by conventional methods.
  • The pharmaceutical compositions may further comprise one or more therapeutic agent. Combination treatments may be administered simultaneously, sequentially or separately, by the same or by different routes, or before, during and after surgical or intervention procedures.
  • The compounds for use of the invention may be prepared by any suitable method known in the art and/or by the processes described below. It will also be appreciated that functional groups, such as amino or hydroxyl groups, present in the various compounds described, and which it is desired to retain, may need to be in protected form before any reaction is initiated. In such instances, removal of the protecting group may be the final step in a particular reaction. Suitable protecting groups for such functionality will be apparent to those skilled in the art. For specific details see "Protective Groups in Organic Synthesis", Wiley Interscience, T W Greene, PGM Wuts. Any mixtures of final products or intermediates obtained can be separated on the basis of the physico-chemical differences of the constituents, in known manner, into the pure final products or intermediates, for example by chromatography, distillation, fractional crystallization, or by formation of a salt if appropriate or possible under the circumstances.
  • The compounds for use according to the invention may be prepared by the following or similar processes.
  • Compound 5-[4-[2-(5-(1-hydroxyethyl)-2-pyridinyl)ethoxy]benzyl]-2,4-thiazolidinedione of formula (1) can be prepared according to Scheme 1 (see e.g. J.Med.Chem. 1996, 39(26),5053).
    Figure imgb0008
  • Amongst other routes, a mixture of compounds (2) and (4) or a mixture of compounds (3) and (5) of the invention may be prepared as in Scheme 1 but using enantiomerically pure alcohols (IIa) and (IIb) as starting materials.
  • The intermediates of formula (IIa) and (IIb) can be prepared as single enantiomers from the racemic alcohol (II) by one or more of the following procedures (Scheme 2):
    1. a) HPLC chiral chromatography separation using chiral columns available in the market.
    2. b) Enzymatic resolution treating the isomeric mixture with an enzyme, such as lipase, which will acetylate one of the isomers leaving the other isomer unreacted. The two isomers can then be readily separated.
    3. c) By treating the isomeric mixture with resolving reagents and separating the resulting diastereoisomers by crystallization or by ordinary column chromatography.
    Figure imgb0009
  • An alternative method to prepare intermediates (IIa) and (IIb) as single enantiomers by chiral synthesis, treating a substrate of formula (I) with an appropriate chiral reducing agent known to those skilled in the art.
  • Yet another method to prepare mixtures (c) - comprising compound (2) and (4) - and (d) - comprising compounds (3) and (5) - (scheme 3), includes the resolution of the racemic mixture VIII using the already described methods (chiral HPLC separation, enzymatic resolution, chiral resolution, etc) followed by double bond reduction in each of the enantiomers Villa and Vlllb.
    Figure imgb0010
  • Mixture (b) (comprising compounds of formula (4) and (5)) and mixture (a) (comprising compounds of formula (2) and (3)) of the invention may be prepared by asymmetric hydrogenolysis of a compound of formula VI using for example Rhodium or Iridium catalysts in the presence of chiral ligands as shown on Scheme 4. Chiral reduction of the double bond may also be performed using biocatalysts (e.g. Rhodotorula rubra and Rhodotorula glutinis).
    Figure imgb0011
  • Compounds of formula (2), (3), (4) and (5) may be obtained from mixtures (c) and (d) (Scheme 5) by chiral HPLC separation. Alternatively, the desired enantiomerically pure compounds can be prepared by chiral synthetic procedures known to those skilled in the art (for example: asymmetric hydrogenolysis of the corresponding single isomer of compound VI).
    Figure imgb0012
  • Abbreviations:
    • ACE: Angiotensin-converting enzyme
    • ADME: Absorption, distribution, metabolism and excretion
    • ALS: Amyotrophic lateral sclerosis
    • AMN: Adrenomyeloneuropathy
    • AUC: Area under the curve
    • C57BL/6 mouse : C57 black 6 mouse
    • cALD: cerebral variant of ALD
    • cAMN: Cerebral adrenomyeloneuropathy
    • CD20: B-lymphocyte antigen CD20
    • CD25: the alpha chain of the IL-2 receptor
    • CD52: Cluster of differentiation 52
    • cDNA: Complementary deoxyribonucleic acid
    • Cmax: Peak plasma concentration after administration.
    • COMT: Catechol O-methyltransferase
    • DEAD: Diethyl azodicarboxylate
    • EC50: Half maximal effective concentration
    • hERG: human Ether-à-go-go-Related Gene
    • HPLC: High performance liquid chromatography
    • LLOQ: Lower limit of quantification
    • MAO-B: Monoamine oxidase B
    • mtDNA: mitochondrial deoxyribonucleic acid
    • NMDA: N-Methyl-D-aspartic acid
    • nDNA: nuclear deoxyribonucleic acid
    • NS: Nervous system
    • Ph: phenyl
    • PPARγ: Peroxisome proliferator-activated receptor-gamma
    • qPCR: Quantitative polymerase chain reaction
    • TIA: Transient ischaemic attack
    • Tmax: Time to reach Cmax
    • VSS: Aparent volume of distribution at steady state
    • X-ALD: X-linked adrenoleukodystrophy
  • The following examples support the invention.
  • Example 1: Pharmacokinetic profile and brain distribution
  • Protocol: Pharmacokinetic parameters and brain distribution of 5-(4-(2-(5-(1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione (racemate or stereoisomers) following single oral (4.5 mg/kg) and intravenous (1 mg/kg) dose administration to male C57 BL/6 mice were determined. Blood samples and brain samples were collected pre-dosing and different times post dosing for both oral and i.v pharmacokinetics. All samples were processed for analysis by protein precipitation using acetonitrile and analyzed with fit-for-purpose LC/MS/MS method. The lower limit of quantification (LLOQ) in plasma and brain for 5-(4-(2-(5-(1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione (1) is 0.99 ng/mL. Pharmacokinetic parameters were calculated using the non-compartmental analysis tool of Phoenix WinNonlin.
  • The results from these experiments are shown in FIG. 1, FIG. 2 and FIG. 3. The data clearly demonstrates that 5-(4-(2-(5-(1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)-thiazolidine-2,4-dione (1) exhibits a good pharmacokinetic profile, low systemic plasma clearance and acceptable Volume of Distribution (Vss) with a brain-plasma ratio exposure of 0.12.
  • Following a single intravenous administration of racemic 5-(4-(2-(5-(1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione (FIG. 1) to C57BL/6 mice at 1 mg/kg dose, the compound exhibited low systemic plasma clearance (11.79 mL/min/kg, normal liver blood flow in mice = 90 mL/min/kg) with terminal elimination plasma half-life of 1.79 hr. The Vss was 2-fold higher than the normal volume of total body water (0.7 L/kg).
  • After a single oral administration of racemic 5-(4-(2-(5-(1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione (1) to C57BL/6 mice at 4.5 mg/kg dose (FIG. 2), plasma concentrations were observed until 24 hr (1 animal). The Tmax in plasma was 0.50 hr. The oral bioavailability was 85%.
  • FIG. 3 shows that brain concentrations for both intravenous and oral pharmacokinetic profiles were observed until 8 hr. Tmax in brain is 0.50 hr with brain-to-plasma exposure (AUClast) ratio of 0.12.
  • These results indicate that 5-(4-(2-(5-(1-hydroxyethyl)pyridine-2-yl)ethoxy) benzyl)thiazolidine-2,4-dione has a favourable pharmacokinetic profile including good oral bioavailability and a brain-to-plasma exposure ratio of 0.12, thus the compound meaningfully crosses the blood brain barrier.
  • Example 2: Mechanism of action: in vitro pharmacology
  • Protocol: To determine the mechanism of action through the agonism of PPAR gamma, a cellular functional assay was performed using a human recombinant cell line cotransfected with a PPRE luciferase reporter, PPAR-y, RXR-α and coactivator DRIP205.
  • Transfected cells were treated with increasing doses of compounds. Luciferase activity was detected by alphascreen technology and normalized based on β-galactosidase activity. The results are expressed as the fold induction over the control (Rosiglitazone 10 µM). Dose response curves were obtained. Results were calculated as EC50 that is the concentration of compound that provokes 50 % control agonist response.
  • EC50 racemic 5-(4-(2-(5-(1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione= 9.3 µM
    The results from these experiments indicate that racemic 5-(4-(2-(5-(1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione and its stereoisomers have varying PPAR gamma agonists activities, with a range of EC50s. These data show that these compounds activate PPAR gamma receptors and consequently the biological functions depending on this activation.
  • Example 3 General experimental conditions:
  • 1H-NMR spectra were recorded on 400 MHz Varian NMR spectrometer using appropriate deuterated solvents. Chromatographic analyses of the compounds were conducted using appropriate methods as shown below.
  • LCMS Method
  • Column: Agilent Zorbax 3.5 µm, SB-C8 (4.6 × 75 mm); wavelength: 210 nm; flow: 1 mL/min; run time: 7 min; mobile phase-gradient (t/%B): 0/30, 3.5/95, 5/95, 5.5/30, 7/30 [A: Water (0.1% formic acid); B: Acetonitrile]; MASS: Agilent-single quad-multimode-APCI-ESI.
  • Chiral HPLC Method
  • Column: Chiralpak-IA 5 µm (4.6 mm x 250 mm); wavelength: 210 nm; flow: 0.7 mL/min; run time: 30 min; mobile phase-isocratic: 65/35 (A/B) [A: n-Hexane (0.05% triethylamine and 0.1% trifluroacetic acid), B: Isopropyl alcohol].
  • Chiral Prep-HPLC Method
  • Column: Chiralpak-IA 5 µm (250 × 20mm); wavelength: 254 nm; flow: 18 ml/min; run time: 60 min; mobile phase-isocratic 50/50 (A/B): A: n-Hexane, B: EtOH (0.05% triehylamine).
  • HPLC Method
  • Column: Symmetry Shield RP-18, 5 µm (4.6 x 250 mm); wavelength: 210 nm; flow: 1mL/min; run time: 28 min; mobile phase-gradient: (t/%B): 0/10, 8/10, 12/60, 16/80, 20/80, 24/10, 28/10 [A: Water (potassium dihydrogen o-phosphate (pH∼3)), B: Acetonitrile]
  • Example 4: 5-(4-(2-(5-(1-hydroxyethyl)pyridin-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione (1) was prepared according to Scheme 6.
  • Figure imgb0013
  • 1-(6-methyl-pyridin-3-yl)-ethanol (II)
  • LiHMDS (1.0 M in tetrahydrofurane, 463 ml, 0.463 mol) was added drop wise to a cooled solution of methyl 6-methylnicotinate (20 g, 0.132 mol) and ethyl acetate (82 g, 0.927 mol) in dimethylformamide at -50°C; gradually raised the temperature to r.t. and stirred at the same temperature. After 1 h, the reaction mixture was cooled to 0°C; slowly diluted with 20% sulphuric acid and heated to reflux. After 4 h, the reaction mixture was cooled to r.t. and further to 0°C and basified with potassium carbonate. The reaction medium was diluted with water and extracted in ethyl acetate (3x50 ml). Combined organic extract was dried over sodium sulphate and concentrated to afford crude 1-(6-methylpyridin-3-yl)ethan-1-one (compound I) (20.0 g) which was taken to next step without any purification.
    ES-MS [M+1]+: 136.1
  • Sodium borohydride (2.3 g, 0.06 mol) was added in small portions over 30 min, to a solution of compound I (16.4 g, 0.121 mol) in ethanol (160 ml) at 0°C and the reaction mixture was stirred at same temperature. After 1 h, the reaction mixture was diluted with sodium bicarbonate solution (sat) (2x200 ml) and extracted with dichloromethane (2x500 ml). The combined organic extract was dried over anhydrous sodium sulphate and concentrated to afford a pale yellow oil, which was purified by flash column chromatography (5% methanol/dichloromethane) to afford compound II (17.0 g; 93% yield over 2 steps) as a pale yellow oil.
    ES-MS [M+1]+: 138.1
    1H NMR (400 MHz, CDCl3): δ 8.35 (d, J = 2.0 Hz, 1H), 7.63 (dd, J = 8.0, 2.4 Hz, 1H), 7.12 (d, J = 8.0 Hz, 1H), 4.89 (q, J = 6.5 Hz, 1H), 3.30 (br s, 1H), 2.50 (s, 3H), 1.48 (d, J = 6.5 Hz, 3H)
  • 5-(1-methoxymethoxy-ethyl)-2-methyl-pyridine (III)
  • Compound II (15 g, 0.109 mol) was added, drop wise, to a cooled suspension of sodium hydride (6.56 g, 0.164 mol) in tetrahydrofurane (150 ml) and stirred at 0°C. After 30 min, chloromethyl methyl ether (13.2 g, 0.164 mol) was added drop wise while stirring and keeping the internal temperature around 0°C. After addition is over, the reaction mixture was stirred at the same temperature for 1 h. The reaction was quenched with ice cold water (80 ml) and extracted with ethyl acetate (3x50 ml). The combined organic extract was dried over anhydrous sodium sulphate and concentrated to afford an orange color oil, which was purified by flash column chromatography (1% methanol/dichloromethane) to afford compound III (10.0 g; 51% yield) as a pale yellow oil.
    ES-MS [M+1]+: 182.2
    1H NMR (400 MHz, CDCl3): δ 8.45 (d, J = 2.0 Hz, 1H), 7.56 (dd, J = 8.0, 2.0 Hz, 1H), 7.14 (d, J = 8.0 Hz, 1H), 4.75 (q, J = 6.4 Hz, 1H), 4.57 (ABq, 2H), 3.36 (s, 3H), 2.53 (s, 3H), 1.48 (d, J = 6.6 Hz, 3H)
  • 2-[5-(1-methoxymethoxy-ethyl)-pyridin-2-yl]-ethanol (IV)
  • A mixture of compound III (7.0 g, 0.0386 mol) and 37% formaldehyde solution (5.8 g, 0.077 mol) was heated to 160°C in a sealed glass tube for 5 h. The reaction mixture was cooled to r.t. and concentrated under reduced pressure to afford a crude compound which was purified by flash column chromatography (1% methanol/dichloromethane) to afford compound IV (1.2 g; 17% yield) as pale yellow oil.
    ES-MS [M+1]+: 212.1
    1H NMR (400 MHz, CDCl3): δ 8.42 (d, J = 2.0 Hz, 1H), 7.65 (dd, J = 8.0, 2.4 Hz, 1H), 7.25 (d, J = 8.0 Hz, 1H), 4.72 (q, J = 6.6 Hz, 1H), 4.65 (t, J = 5.6 Hz, 1H), 4.52 (ABq, 2H), 3.73 (m, 2H), 3.24 (s, 3H), 2.86 (t, J = 7.2 Hz, 2H), 1.49 (d, J = 6.4 Hz, 3H).
  • 4-{2-[5-(1-methoxymethoxy-ethyl)-pyridin-2-yl]-ethoxy}-benzaldehyde (V)
  • Methanesulphonylchloride (1.19 g, 0.01 mol) was added, drop wise, to a cooled suspension of compound IV (1.7 g, 0.008 mol) and triethylamine (1.79 ml, 0.013 mol) in dichloromethane (20 ml) at 0°C and stirred at same temperature for 1 h. The reaction mixture was diluted with water (50 ml) and extracted with dichloromethane (3x50 ml). The combined organic extract was dried over anhydrous sodium sulphate and concentrated to afford 2-(5-(1-(methoxymethoxy)ethyl)pyridin-2-yl)ethyl methanesulfonate (2.04 g; 88% yield) as a yellow oil, which was taken to next step without purification.
    ES-MS [M+1]+: 290
    2-(5-(1-(methoxymethoxy)ethyl)pyridin-2-yl)ethyl methanesulfonate was added (2.3 g, 0.008 mol) to a stirred suspension of 4-hydroxybenzaldehyde (1.65 g, 0.0137 mol) and potassium carbonate (1.86 g, 0.0137 mol) in mixture of toluene (25 ml) and ethanol (25 ml); stirred at 85°C for 5 h. After consumption of the starting materials, the reaction mixture was diluted with water (30 ml) and extracted with ethyl acetate (2x100 ml). The combined organic extract was washed with water; dried over anhydrous sodium sulphate and concentrated to afford a crude dark yellow liquid. The crude was purified by flash column chromatography (1% methanol/dichloromethane) to afford compound V (1.5 g; 60% yield) as pale yellow liquid.
    ES-MS [M+1]+: 316.1
  • 5-(4-{2-[5-(1-methoxymethoxy-ethyl)-pyridin-2-yl]-ethoxy}-benzylidene)-thiazolidine-2,4-dione (VI)
  • Piperidine (80 mg, 0.95 mmol) was added to a solution of compound V (0.6 g, 1.9 mmol) and thiazolidine-2,4-dione (0.22 g, 1.9 mmol) in ethanol (15 ml) and the mixture was heated to reflux overnight. After 15 h, the reaction mixture was cooled to r.t. and concentrated under reduced pressure to afford crude mixture, which was purified by flash column chromatography (2% methanol/dichloromethane) to afford compound VI (500 mg; 64% yield) as a yellow solid.
    ES-MS [M+1]+: 415.1
    1H NMR (400 MHz, DMSO-d6): δ 12.25 (br s, 1H), 8.47 (d, J = 2.0 Hz, 1H), 7.70 (dd, J = 8.0, 2.0 Hz, 1H), 7.54 (d, J = 8.8 Hz, 2H), 7.36 (d, J = 8.0 Hz, 1H), 7.21 (d, J = 8.8 Hz, 2H), 4.73 (m, 1H), 4.60-4.40 (m, 4H), 4.22 (t, J = 6.2 Hz, 1H), 3.24 (s, 3H), 3.20 (t, J = 6.8 Hz, 2H), 1.41 (d, J = 6.0 Hz, 3H).
  • 5-(4-{2-[5-(1-hydroxy-ethyl)-pyridin-2-yl]-ethoxy}-benzyl)-thiazolidine-2,4-dione (1)
  • A solution of sodium borohydride (115 mg, 3.017 mmol) in 0.2N sodium hydroxide (1.2 ml) was added slowly to a stirred solution of compound VI (0.5 g, 1.207 mmol), dimethylglyoxime (42 mg, 0.36 mmol) and COCl2.6H2O (23 mg, 0.096 mmol) in a mixture of water (6 ml): tetrahydrofurane (6 ml) and 1M sodium hydroxide (1 ml) solution at 10°C and after addition, the reaction mixture was stirred at r.t.. After 1 h, the reaction color lightened and additional quantities of sodium borohydride (46 mg, 1.207 mmol) and CoCl2.6H2O (22 mg, 0.096 mmol) were added and stirring was continued at r.t. After 12 h, the reaction was neutralized with acetic acid (pH∼7); diluted with water (10 ml) and extracted in ethyl acetate (3x50 ml). The combined organic extract was dried over anhydrous sodium sulphate and concentrated to afford crude compound VII, 5-(4-(2-(5-(1-(methoxymethoxy)ethyl)pyridin-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione (0.4 g) as pale yellow semi solid, which was taken to next step without purification.
    ES-MS [M+1]+: 417.5
    2N HCI (2 ml) was added to a solution of compound VII (0.4 g, 0.96 mmol) in methanol (20 ml) and the mixture was heated to reflux. After 4 h, the reaction mixture was cooled to r.t.; concentrated under reduced pressure to afford a residue which was dissolved in water and the solution was neutralized using sodium bicarbonate solution (sat). The resulting white precipitate was collected by filtration to afford compound 1 (250 mg; 56% yield over 2 steps) as an off-white solid.
    ES-MS [M+1]+: 373.4
    1H NMR (400 MHz, DMSO-d6): δ 12.00 (br s, -NH), 8.46 (d, J = 2.0 Hz, 1H), 7.66 (dd, J = 8.0, 2.4 Hz, 1H), 7.30 (d, J = 8.0 Hz, 1H), 7.13 (d, J = 8.4 Hz, 2H), 6.86 (d, J = 8.4 Hz, 2H), 5.25 (d, J = 4.4 Hz, 1H), 4.86 (m, 1H), 4.75 (m, 1H), 4.30 (t, J = 6.8 Hz, 2H), 3.30 (m, 1H), 3.14 (t, J = 6.4 Hz, 2H), 3.04 (m, 1H), 1.34 (d, J = 6.4 Hz, 3H).
  • Example 5 (mixture (c) of compounds (2) and (4)) and (mixture (c) of compounds (3) and (4))
  • A mixture of compounds (2) and (4) (mixture (c)) and a mixture of compounds (3) and (5) (mixture (d)) were prepared according to Scheme 7.
    Figure imgb0014
  • The methyl chloromethyl ether group of (Z)-5-(4-(2-(5-(1-(methoxy-methoxy)ethyl)pyridin-2-yl)ethoxy)benzylidene)thiazolidine-2,4-dione (compound VI) was removed by treatment with aqueous HCI to give the racemic alcohol VIII.
  • The enantiomers contained in the racemic mixture of (Z)-5-(4-(2-(5-(1-hydroxyethyl)pyridin-2-yl)ethoxy)benzylidene)thiazolidine-2,4-dione (VIII) were separated by HPLC chiral chromatography to yield (R)-VIII and (S)-VIII..
  • (R)-VIII was then treated with a reducing mixture (CoCl2-6H2O, dimethylglyoxime, NaOH, sodium borohydride), (modified conjugate reduction protocol of Pfaltz), to yield mixture (c) comprising compounds (2) and (4).
  • (S)-VIII was then treated with a reducing mixture (CoCl2-6H2O, dimethylglyoxime, NaOH, sodium borohydride), (modified conjugate reduction protocol of Pfaltz), to yield mixture (d) comprising compounds (3) and (5).
  • Example 6: Preparation of diastereomeric mixtures D-1 and D-2 of M-IV:
  • Figure imgb0015
  • Step 1: Synthesis of compound VIII: HCl (48 ml, 2N) was added to a solution of compound VI (10 g, 0.024 mol) in methanol (200 ml) and the mixture was heated to reflux. After 4 h of reflux, the reaction mixture was cooled to r.t. and concentrated under reduced pressure to afford a yellow solid. The solid was suspended in water (70 ml) and neutralized using a saturated NaHCO3 solution. The resulting pale yellow precipitate was collected by filtration and vacuum dried to afford compound VIII (7.5 g; 84% yield).
    ES-MS [M+1]+: 371.0.
  • Step 2: Chiral prep. HPLC
  • Compound VIII (1.0 g) was dissolved in a mixture containing equal volumes of acetonitrile, methanol and dichloromethane; injected (150 µl injections) in chiral prep-HPLC column (Chiralpak-IA 250 x 20 mm, 5 micron) and separated [Mobile phase- n-Hexane/0.05% Et3N in EtOH (50:50); flow Rate: 18ml/min; run time: 60 min]. The fractions containing the enantiomers Villa and Vlllb were separately concentrated under reduced pressure to minimum volume and the respective residues were diluted with EtOAc (100 ml), followed by water (50 ml). The resultant organic phases were dried over anhydrous Na2SO4 and concentrated to afford compounds VIIIa and VIIIb as off-white solids. Enantiomers VIIIa and VIIIb were isolated but the absolute configuration of each enantiomer has not been determined.
  • Compound Ent-1 (VIII): 250 mg (Yield: 50%); tR (Chiral HPLC) = 14.8 min; ES-MS [M+1]+: 371.0; 1H NMR (400 MHz, DMSO-d6): δ 12.5 (br S, 1H), 8.47 (s, 1H), 7.71 (s, 1H), 7.67 (dd, J = 8.0, 2.0 Hz, 1H), 7.53 (d , J = 9.2 Hz, 2H), 7.31 (d, J = 7.6 Hz, 1H), 7.08 (d, J = 8.8 Hz, 2H), 5.25 (d, J = 4.0 Hz, 1H), 4.74-4.76 (m, 1H), 4.43 (dd, J = 6.8, 6.4 Hz, 2H), 3.18 (t, J = 6.4 Hz, 2H), 1.34 (d, J = 6.4 Hz, 3H).
  • Compound Ent-2 (VIII): 237 mg (Yield: 47%); tR (Chiral HPLC) = 16.7 min; ES-MS [M+1]+: 371.0; 1H NMR (400 MHz, DMSO-d6): δ 12.5 (br S, 1H), 8.47 (s, 1H), 7.71 (s, 1H), 7.67 (dd, J = 8.0, 2.0 Hz, 1H), 7.53 (d , J = 8.8 Hz, 2H), 7.31 (d, J = 8.0 Hz, 1H), 7.08 (d, J = 9.2 Hz, 2H), 5.23 (d, J = 3.6Hz, 1H), 4.75 (m, 1H), 4.43 (dd, J = 6.8, 6.4 Hz, 2H), 3.18 (dd, J = 6.8, 6.4 Hz, 2H), 1.34 (d, J = 6.4 Hz, 3H).
  • Synthesis of diastereomeric mixtures of M-IV Synthesis of D-1 MIV
  • Step 3: A solution of NaBH4 (77 mg, 2.02 mmol) in 0.1 N NaOH (2 ml) was added slowly to a stirred solution of compound Ent-1 (VIII) (250 mg, 0.675 mmol), dimethylglyoxime (32 mg, 0.27 mmol) and CoCl2.6H2O (16 mg, 0.067 mmol) in a mixture of water (10 ml), THF (10 ml) and 1M NaOH (0.5ml) solution at 10 °C, and the reaction mixture was stirred at r.t. for 1 h. After color of the reaction medium faded, additional quantity of NaBH4 (26 mg, 0.675 mmol) and CoCl2.6H2O (16 mg, 0.067 mmol) were added and stirring was continued at r.t. [additional quantities of CoCl2 and NaBH4 were added at 12 h intervals till the starting material was consumed, as monitored by LCMS]. After 90-96 h, the reaction mixture was neutralized with AcOH (pH∼7); diluted with water (10 ml) and extracted in EtOAc (3 × 50 ml). The combined organic extract was dried over anhydrous Na2SO4 and concentrated to afford crude compound which was purified by flash column chromatography (SiO2; 4% methanol in CH2Cl2) to afford diastereomeric mixture of MIV D-1 (125 mg) as off-white solid.
  • Synthesis of D-2 MIV
  • Step 3: A solution of NaBH4 (72 mg, 1.921 mmol) in 0.1 N NaOH (2 ml) was added slowly to a stirred solution of compound Ent-2 (VIII) (237 mg, 0.64 mmol), dimethylglyoxime (30 mg, 0.256 mmol) and COCl2.6H2O (15 mg, 0.064 mmol) in a mixture of water (10 ml), THF (10 ml), and 1M NaOH (0.5ml) solution at 10 °C, and the reaction mixture was stirred at r.t. for 1 h. After color of the reaction medium faded, additional quantity of NaBH4 (24 mg, 0.64 mmol) and CoCl2.6H2O (15 mg, 0.064 mmol) were added and stirring was continued at r.t. [additional quantities of CoCl2.6H2O and NaBH4 were added at 12 h intervals till the starting material was consumed, as monitored by LCMS]. After 96 h, the reaction mixture was neutralized with AcOH (pH∼7); diluted with water (10 ml) and extracted in EtOAc (3 x 50 ml). The combined organic extract was dried over anhydrous Na2SO4 and concentrated to afford crude compound, which was purified by flash column chromatography (SiO2; 4% methanol in CH2Cl2) to afford diastereomeric mixture of MIV D-2 (100 mg) as off-white solid.
  • MIV D-1: yield: 125 mg (50%); tR (Chiral HPLC) = 17.8, 14.7 min; ES-MS [M+1]+: 373.0, 1H NMR (400 MHz, DMSO-d 6): δ 12.00 (br s, NH), 8.46 (d, J = 2.0 Hz, 1H), 7.67 (dd, J = 8.0, 2.4 Hz, 1H), 7.30 (d, J = 8.0 Hz, 1H), 7.13 (d, J = 8.8Hz, 2H), 6.86 (d, J = 8.4 Hz, 2H), 5.27 (d, J = 4.0 Hz, 1H), 4.88-4.85 (m, 1H), 4.76-4.74 (m, 1H), 4.30 (t, J = 6.8 Hz, 2H), 3.30 (m, 1H), 3.14 (dd, J = 6.8, 6.4 Hz, 2H), 3.08-3.02 (m, 1H), 1.34 (d, J = 6.4 Hz, 3H).
  • MIV D-2: yield: 100 mg (42%); tR (Chiral HPLC) = 19.4, 16.5 min; ES-MS [M+1]+: 373.0; 1H NMR (400 MHz, DMSO-d 6): δ 12.01 (br s, -NH), (d, J = 2.0 Hz, 1H), 7.67 (dd, J = 8.0, 2.0 Hz, 1H), 7.31 (d, J = 8.0 Hz, 1H), 7.13 (d, J = 8.8 Hz, 2H), 6.86 (d, J = 8.8 Hz, 2H), 5.27 (d, J = 4.0 Hz, 1H), 4.88-4.85 (m, 1H), 4.76-4.74 (m, 1H), 4.30 (dd, J = 6.8, 6.4 Hz, 2H), 3.30 (m, 1H), 3.14 (dd, J = 6.8, 6.4 Hz, 2H), 3.08-3.02 (m, 1H), 1.34 (d, J = 6.8 Hz, 3H).
  • Diastereomeric mixtures D-1 and D-2 of MIV correspond to mixtures (c) and (d) described above, but the specific diastereomers present in each diastereomeric mixture have not been assigned.
  • Example 7: in vitro ADME and toxicological characterization
  • Protocol: The assays performed include cytochrome P450 inhibition with the different isoforms, microsomal and hepatocyte stability, neurotoxicity in neural cells and hERG safety assays using a patch clamp electrophysiology measurement (FDA Draft Guidance for Industry. Drug Interaction Studies - Study Design, Data Analysis, Implications for Dosing, and Labelling Recommendations 2012, The European Medicines Agency (EMA) Guideline on the Investigation of Drug Interactions Adopted in 2012, Schroeder K et al. 2003 J Biomol Screen 8 (1); 50-64, Barter ZE et al. 2007 Curr Drug Metab 8 (1); 33-45, LeCluyse EL and Alexandre E 2010 Methods Mol Biol 640; 57-82). The results indicate a safe and favourable ADME profile for the compounds of the invention.
  • Example 8: The brain plasma ratios of Pioglitazone, MIV, Mill and Mil following oral dosing of a single administration of Pioglitazone at 4.5 mg/kg in male C57BL/6 mice.
  • The brain-plasma ratio was calculated based on levels of Pioglitazone, MIV, Mill and Mllin plasma and brain quantified at C max (maximal concentration) following oral dosing of a single administration of Pioglitazone at 4.5 mg/kg in male C57BL/6 mice.
  • The percentage brain plasma ratio was 9, 13, 7 and 1 %, respectively, for Pioglitazone, MIV, Mil and Mill as shown in the Figure 4. Thus, active metabolites Mill and Mil crossed the BBB at much lower extent than Pioglitazone as it was predicted based on the physicochemical properties of the compounds (see Table1). In contrast, unexpectedly metabolite MIV crossed the BBB in a higher percentage than the parent compound Piolgitazone
  • The calculations of the both indexes (ClogP and QPlogBB) for Pioglitazone and its metabolites Mil and Mill are shown in Table 1. For both indexes the 2 metabolites are lower than for pioglitazone, suggesting for MII, and MIII a less favored penetration and distribution within CNS. TABLE 1
    Structure Name QPlogBB CLogP HBD HBA
    Figure imgb0016
    Pioglitazone -1.22 3.53 1 4
    Figure imgb0017
    MIV -1.72 1.78 2 5
    Figure imgb0018
    MIII -1.66 2.34 1 5
    Figure imgb0019
    MII -1.72 2.13 2 5
  • Example 9: The brain plasma ratios of Pioglitazone and MIV and following oral dosing of a single administration of either Pioglitazone or M-IV both at 4.5 mg/kg in male C57BL/6 mice.
  • In order to confirm the findings showed in the last example, additional experiments were done. The brain-plasma ratio was calculated based on the pharmacokinetics curves of plasma and brain concentration-time profiles calculated as area under the curves of Pioglitazone and following oral dosing of a single administration of either Pioglitazone or MIV both at 4.5 mg/kg in male C57BL/6 mice.
  • The percentage brain-plasma ratio was 8 % and 12% for Pioglitazone and M4 respectively as shown in the Figure 5. This 50% of increase in brain -plasma ratio for the hydroxilated metabolite M-IV compared with that the one observed with Pioglitazone under the same condition, was totally unexpected based on the on the physicochemical properties predictive calculations (see Table 1).
  • M-IV shows a behavior contrary to that expected. As Mil, MIV is a monohydroxylated metabolite, but instead of decreasing around 50% its BBB penetration, the BBB penetration is 50% higher.
  • The calculations of the both indexes (ClogP and QPlogBB) for Pioglitazone and M-IV are shown in Table 1. For both indexes MIV shows a lower value than pioglitazone, suggesting for M-IV a less favored penetration and distribution within CNS, contrary to what has surprisingly been observed experimentally.
  • Example 10: Characterization of In vivo epimerization of the two diastereomeric mixtures of MIV, D-1 and D-2 in mice.
  • Protocol: Pharmacokinetic parameters of diastereomeric mixtures D-1 and D-2 of 5-(4-(2-(5-(1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione following single oral (4.5 mg/kg) gavage dose administration to male Swiss albino mice were determined. A total of 51 mice were used in this study with parallel sampling design (n=3/time point). Blood samples and were collected pre-dosing and different times post dosing for both oral pharmacokinetics.
  • Diastereomeric mixtures D-1 and D-2 of 5-(4-(2-(5-(1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione were extracted from Swiss albino mouse plasma samples using liquid-liquid extraction (LLE) method and quantified using liquid chromatography tandem mass spectrometry (LC-MS/MS) with Electro Spray Ionization (ESI) and multiple reaction monitoring (MRM). Selected plasma and brain samples were subjected for the chiral analysis using Chiral AGP column to identify the chiral inter-conversion. Achiral bioanalytical method was employed to quantify the total M-IV present in the plasma and brain samples.
  • Formulation samples were suitably diluted with 70% methanol and the instrument response was compared against known corresponding diastereomeric mixture standard using achiral LC-MS/MS method. The lower limit of quantification (LLOQ) in plasma for diastereomeric mixture D-1 and D-2 of 5-(4-(2-(5-(1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione (1) is 0.99 ng/mL. Pharmacokinetic parameters were calculated using the non-compartmental analysis tool of Phoenix WinNonlin.
  • The results from these experiments are shown in FIG 6. The data clearly demonstrates that the difference in % of conversion among diastereomeric mixtures is high in mice. In vivo, both D-1 and D-2 interconvert, although the conversion to D-2 from D-1 is much more accentuated than the conversion from D-1 to D-2.
  • Example 11: Characterization of cortical neurons glutamate injured as a model of Alzheimer's Disease.
  • Primary cortical neurons injured by glutamate (excitotoxicity) are a well established in vitro model for neurodegenerative disorders ( J Neurosci; 1999 Apr 1;19(7):2455-63 ; J Neurosci Res. 2013 May;91(5):706-16 ) such as Parkinson's disease, Alzheimer's disease, and Huntington's disease (Brain Res Bull 2013 Apr;93:27-31.), but also in other pathologies such as multiple sclerosis (Scand J Immunol 2014; 79(3):181-186 ).
  • Protocol: Rat cortical neurons were cultured as described by Singer (J Neurosci. 1999 Apr 1;19(7):2455-63) and Callizot (J Neurosci Res. 2013 May;91(5):706-16).
  • Foetuses were collected and immediately placed in ice-cold Leibovitz medium. Cortex was treated with a trypsin- EDTA solution and the dissociation was stopped by addition of Dulbecco's modified Eagle's medium (DMEM) with glucose (Pan Biotech), containing DNAse I and 10% fetal calf serum (FCS). Cells were mechanically dissociated and centrifuged. The pellet was resuspended in a defined culture medium with 10 ng/ml of brain-derived neurotrophic factor (BDNF). The cells were seeded in 96-well plates precoated with poly-L-lysine and were cultured at 37°. The medium was changed every 2 days. The cortical neurons were intoxicated with glutamate after 13 days of culture.
  • Briefly, on day 13 of culture, BDNF and test compound were pre-incubated with primary cortical neurons for 1 hour before glutamate exposure. Glutamate was added to a final concentration of 40 µM diluted in control medium in presence of BDNF or test compound for 20 min.
  • After 20 min, glutamate was washed out and fresh culture medium with BDNF or test compound was added for additional 48 hours. The survival evaluation was done by a MTT assay performed with CellTiter 96® Aqueous One Solution Cell Proliferation Assay (Promega).
  • The results are shown in FIG 7. They show that on a glutamate injury, MIV (compound (1)) shows a protective effect (reaching the significance for the 3 µM). Interestingly we observed a nice bell shape curve for MIV. At 3 µM we have a full protective effect as the one observed with the reference compound (BDNF 50 ng/ml). All values are expressed as mean +/- SE. Statistical analysis was performed by one-way ANOVA, followed by Dunnett's or PLSD Fisher test, p < 0.05 are considered significant.
  • Example 12: Characterization of inhibition of MAO B (Monoamine oxidases) as a potential drug for treating Parkinson's Disease.
  • Selective inhibitors MAO-B increase dopamine levels in the CNS affected in Parkinson's disease without increasing levels of the other neurotransmisors (epinephrine, norepinephrine or serotonine), in contrast to no selective MAO inhibitors (MAO-A and MAO B). The MAO-B inhibitors can be used also to treat depressions.
  • Protocol: Human recombinant monoamine oxidase proteins MAO-A and MAO-B were purchased from Sigma Aldrich (Reference M7316 and M7441 respectively). In order to monitor the MAO enzymatic activities and their inhibition rate a fluorescence based assay was used. The substrate for the assay, kynuramine, is non-fluorescent until undergoing oxidative deamination by MAOs resulting in the fluorescent product 4-hydroxyquinoline. Kynuramine is a substrate for both MAO-A and -B (non-specific substrate). Clorgiline and Deprenyl (Sigma Aldrich) were used as controls for specific inhibition of MAO-A and MAO-B respectively.
  • Results show that 5-(4-(2-(5-(1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione inhibits MAO B with a IC50 of 70.5 nM. In contrast, this compound did not inhibit MAO A protein.
  • Example 13: Characterization of in vivo efficacy in an animal model experimental autoimmune encephalomyelitis (EAE) as a model of neuroinflammatory diseases.
  • Neuroinflammation can be initiated in response to a variety of infection, traumatic brain injury, toxics, or autoimmunity in the CNS.
  • The neuroinflammatory models are characterized by proliferation of astrocytes and microglia, along with neuronal loss, is a prominent feature of many diseases of the central nervous system, including Alzheimer's Disease, Multiple Sclerosis, stroke, Parkinson, traumatic brain injury, infection and ALD (Human Molecular Genetics, 2004, Vol. 13, No. 23 2997-3006).
  • Chronic inflammation is the sustained activation of glial cells and recruitment of other immune cells into the brain. It is chronic inflammation that is typically associated with neurodegenerative diseases.
  • The EAE model is a neuroinflammatory model, classically used for multiple sclerosis, which resembles and includes most of the features of the severe cerebral forms of ALD, microglial activation, brain demyelination and axonal degeneration as well. Although the ethiology of the disease is different from ALD and the EAE (a model of multiple sclerosis triggered by autoreactive CD4+ lymphocytes), the EAE model is a valuable tool for studying therapies for both ALD and multiple sclerosis (Nature 2007; 7:904-912).
  • Protocol: The development of clinical symptoms in multiple sclerosis and its animal model experimental autoimmune encephalomyelitis (EAE) involves T-cell activation and migration into the central nervous system, production of glial-derived inflammatory molecules, and demyelination and axonal damage. Chronic, monophasic EAE was actively induced as described with greater than 98% pure synthetic myelin oligodendrocyte glycoprotein peptide 35-55 (MOG35-55, MEVGWYRSPFSRWHLYRNGK, SEQ ID NO: 1). Female C57BL/6 mice (6-8 weeks old) were injected (two 100 µL subcutaneous injections into one hind limb) with 250 µg of MOG35-55 emulsified in a 100 µL phosphate-buffered solution mixed with 100 uL of complete Freund's adjuvant containing 500 µg of Mycobacterium tuberculosis (Difco, Detroit, MI). The mice received an injection of pertussis toxin (400 ng in 200 µL of a phosphate-buffered solution, intraperitoneally), a second pertussis toxin injection 2 days later, and a booster injection of MOG35-55 at 7 days. Clinical signs were scored as follows: 0, no signs; 1.0, limp tail/loss of righting; 2.0, ataxia with limp tail; 3.0, paralysis of single hind limb; 4.0, paralysis of both hind limbs; 4.5, moribund; and 5.0, death. For both models, scores of 5 were noted and counted on the day of death only.
  • The compound was administered twice a day (bid) started on day 5 post immunization for 15 days at three different increasing doses.
  • The results showed that MIV (compound (1)) reduces the development and severity of experimental autoimmune encephalomyelitis model. Average daily clinical scores from the experiment are shown in FIG 9. The clinical symptoms decreases in a dose-dependent manner, the highest doses show the maximal effect. Clinical symptoms were reduced by MIV, suggesting a role for PPARgamma activation in protective effects. No body weight loss and no significant hematological toxicity at highest doses associated to treatment.
  • Neuroinflammation is a hallmark of both multiple sclerosis and ALD, thus MIV may be effective on both diseases.. In fact, decreasing microglia activation provides a molecular basis for explaining why allogeneic and autologous HSCT are effective at arresting the cerebral inflammation, namely by the replacement and the functional metabolic restoration of the monocyte lineage and connect cALD and AMN phenotypes with a shared pathogenic pathway (Human Molecular Genetics, 2012, Vol. 21, No. 5 1062-1077). Thus, these models may be have a great potential and relevance to study the role of PPAR gamma agonists in ALD.
  • Example 14: Characterization in fibroblasts from patients of X-ALD as a model of X-linked adrenoleukodystrophy.
  • Human control and X-linked adrenoleukodystrophy fibroblasts were obtained from Coriell (Candem, USA). Cells were grown in Dulbecco's modified Eagle medium containing 10% foetal bovine serum, 100 U/ml penicillin and 100 mg streptomycin, at 37 °C in humidified 95% air/5% CO2, to 80-90% confluence. To perform our experiments, Dulbecco's modified Eagle medium without D-glucose, pyruvate or L-glutamine was used. Cells were cultured in this medium supplemented with 1 g/l of glucose or 1 g/l of galactose and 10% foetal bovine serum for 24 hours incubated with increasing doses of MIV (3, 10 and 30 µM).
  • The determination of MTT was performed as described by Mosmann J. Immunol. Methods 1983, 65,55-63 and by Hansen J. Immunol. Methods 1989; 119,203-210. This method is based on the ability of viable but not dead cells to convert the tetrazolium salt (MTT) to colored formazan.
  • For the determination of ATP levels, 2x104 cells/well were seeded in 96 well cell culture plate in complete medium. After 16-18 h cells, were lysed in 20 µl lysis buffer and 10 µl of lysate was used to measure ATP levels using ATP determination Kit (Molecular Probes, Invitrogen). 1 µl each of the remaining lysate was used for protein measurement.
  • Results show a protective effect of MIV (compound (1)) on ALD fibroblasts based on increase in cell survival (20 % at 3, µM, vs non-treated).
  • Example 15: Characterization of Spinal cord motor neurons as a model of motor neuron diseases (ALS)
  • Spinal cord motor neurons injured by glutamate are an in vitro experimental model suitable for studying ALS and other motor neuron diseases (MNDs) such as Progressive bulbar palsy, pseudobulbar palsy, primary lateral sclerosis (PLS), progressive muscular atrophy, spinal muscular atrophy (SMA), post-polio syndrome (PPS) and other rare diseases such us Charcot-Marie-Tooth disease, Guillain-Barre syndrome or AMN ( Neuron. 1992 Apr;8(4):737-44 ).
  • Protocol: Rat spinal cord (SC) motor neurons were cultured as described by Martinou (Neuron. 1992 Apr;8(4):737-44) and Wang (PLoS Genet. 2013;9(9)). Briefly, pregnant female rats of 14 days gestation were killed by cervical dislocation and foetuses were collected and immediately placed in ice-cold L15 Leibovitz medium. Spinal cord was treated for 20 min at 37°C with a trypsin- EDTA. The dissociation was stopped by addition of Dulbecco's modified Eagle's medium (DMEM) with glucose (Pan Biotech), containing DNAse I and 10% fetal calf serum (FCS). Cells were mechanically dissociated and they were then centrifuged. The pellet was resuspended in a defined culture medium with 10 ng/ml of brain-derived neurotrophic factor (BDNF). The cells were seeded in 96-well plates precoated with poly-L-lysine and were cultured at 37°. The medium was changed every 2 days.
  • Briefly, on day 13 of culture, BDNF or test compound was pre-incubated with primary Spinal cord (SC) motor neurons for 1 hour before glutamate exposure. Glutamate was added to a final concentration of 10 µM diluted in control medium in presence of BDNF or test compound for 20 min. After 20 min, glutamate was washed out and fresh culture medium with BDNF or test compound was added for additional 48 hours.
  • The survival evaluation was done by immunostaining. 48 after the intoxication, the cell culture supernatant was taken off and the SC motor neurons were fixed by a cold solution of ethanol (95%) and acetic acid (5%) for 5 min and permeabilized. Cells were incubated for 2 hours with a monoclonal antibody anti microtubule-associated-protein 2 (MAP-2) that stains specifically cell bodies of neurons (MAP-2) allowing study of neuron survival evaluation in the culture. This antibody was revealed with Alexa Fluor 488 goat anti-mouse IgG).
  • For each condition 6 wells were assessed, 30 pictures per well were taken using ImageXpress (Molecular Device) with 20x magnification. All images were taken with the same conditions. Analysis of total number of neurons was performed automatically by using Custom module editor (Molecular Device).
  • Test compounds were pre-incubated for 1 hour before glutamate application.
  • The results (FIG 10) showed that MIV (compound (1), 1 µM) on a glutamate injury has a protective effect in SC motor neurons (MN), demonstrated by a statistically different (p<0.05 t Student's) increase in cell survival vs the control with glutamate (Glut).
  • SEQUENCE LISTING
    • <110> MINORYX THERAPEUTICS S.L.
    • <120> 2,4-Thiazolidinedione derivatives in the treatment of central nervous system disorders
    • <130> P10708EPPC01
    • <150> EP14382130.4
      <151> 2014-04-02
    • <160> 1
    • <170> PatentIn version 3.5
    • <210> 1
      <211> 21
      <212> PRT
      <213> Artificial Sequence
    • <220>
      <223> myelin oligodendrocyte glycoprotein peptide 35-55
    • <400> 1
      Figure imgb0020

Claims (13)

  1. A compound of formula (1) or a pharmaceutically acceptable salt thereof for use in the treatment or prevention of a central nervous system disorder
    Figure imgb0021
    wherein the central nervous system disorder is selected from the group consisting of Friedreich's ataxia, a brain injury, a rare metabolic disease, global or local ischemia, intracerebral haemorrhage, stroke, vascular dementia, meningitis caused by viral infection in the nervous system, encephalitis caused by viral infection in the nervous system, and a metastasis of a primary brain tumor.
  2. The compound for use according to claim 1 selected from compounds (2) to (5):
    (2) (R)-5-(4-(2-(5-((R)-1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione;
    (3) (R)-5-(4-(2-(5-((S)-1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione;
    (4) (S)-5-(4-(2-(5-((R)-1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione; and
    (5) (S)-5-(4-(2-(5-((S)-1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione;
    or a pharmaceutically acceptable salt thereof.
  3. The compound for use according to claim 1 or claim 2, wherein no more than 0.015 % of the total number of hydrogen atoms per mole of compound are in the form of the 2H isotope.
  4. A mixture of two or more of the compounds selected from compounds (2) to (5):
    (2) (R)-5-(4-(2-(5-((R)-1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione;
    (3) (R)-5-(4-(2-(5-((S)-1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione;
    (4) (S)-5-(4-(2-(5-((R)-1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione; and
    (5) (S)-5-(4-(2-(5-((S)-1-hydroxyethyl)pyridine-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione;
    or a pharmaceutically acceptable salt thereof,
    for use in the treatment or prevention of a central nervous system disorder, wherein the disorder is selected from the group consisting of Friedreich's ataxia, a brain injury, a rare metabolic disease, global or local ischemia, intracerebral haemorrhage, stroke, vascular dementia, meningitis caused by viral infection in the nervous system, encephalitis caused by viral infection in the nervous system, and a metastasis of a primary brain tumor.
  5. The mixture for use according to claim 4 selected from the group consisting of:
    (a) Mixtures comprising compounds (2) and (3);
    (b) Mixtures comprising compounds (4) and (5);
    (c) Mixtures comprising compounds (2) and (4); and
    (d) Mixtures comprising compounds (3) and (5).
  6. The compound or mixture of compounds for use according to any one of claims 1-5, wherein the disorder is Friedreich's ataxia.
  7. The compound or mixture of compounds for use according to any one of claims 1-5, wherein the central nervous system disorder is a rare metabolic disease.
  8. The compound or mixture of compounds for use according to any one of claims 1-7, wherein another therapeutic agent is also administered.
  9. The compound or mixture of compounds for use according to claim 8, wherein the compound or mixture of compounds and said other therapeutic agent are provided in combination.
  10. The compound or mixture of compounds for use according to any one of claims 1-9, wherein the compound or mixture of compounds is provided in a pharmaceutical composition, such as an oral form of the pharmaceutical composition.
  11. The compound or mixture of compounds for use according to claim 10, wherein the oral form is an oral solution or an oral suspension; or the oral form is selected from the group consisting of tablets, capsules, pills, and granules.
  12. The compound or mixture of compounds for use according to any one of claims 9-11, wherein the compound or mixture of compounds in the pharmaceutical composition is at a dosage of from 0.1 mg to 200 mg or from 10 mg to 100 mg.
  13. The compound or mixture of compounds for use according to claim 9, wherein the pharmaceutical composition is suitable for topical, epicutaneous, subcutaneous, transdermal, intramuscular, parenteral, ocular, rectal, vaginal, inhalation, buccal, sublingual, or intranasal delivery, such as a sublingual dosage form.
EP18173541.6A 2014-04-02 2015-04-01 2,4-thiazolidinedione derivatives in the treatment of central nervous system disorders Active EP3388064B1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
SI201531591T SI3388064T1 (en) 2014-04-02 2015-04-01 2,4-thiazolidinedione derivatives in the treatment of central nervous system disorders
PL18173541T PL3388064T3 (en) 2014-04-02 2015-04-01 2,4-thiazolidinedione derivatives in the treatment of central nervous system disorders
HRP20210518TT HRP20210518T1 (en) 2014-04-02 2021-03-30 2,4-thiazolidinedione derivatives in the treatment of central nervous system disorders

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP14382130 2014-04-02
PCT/EP2015/057224 WO2015150476A1 (en) 2014-04-02 2015-04-01 2,4-thiazolidinedione derivatives in the treatment of central nervous system disorders
EP15713499.0A EP3125888B1 (en) 2014-04-02 2015-04-01 2,4-thiazolidinedione derivatives in the treatment of central nervous system disorders

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
EP15713499.0A Division EP3125888B1 (en) 2014-04-02 2015-04-01 2,4-thiazolidinedione derivatives in the treatment of central nervous system disorders

Publications (2)

Publication Number Publication Date
EP3388064A1 EP3388064A1 (en) 2018-10-17
EP3388064B1 true EP3388064B1 (en) 2021-03-10

Family

ID=50513194

Family Applications (2)

Application Number Title Priority Date Filing Date
EP18173541.6A Active EP3388064B1 (en) 2014-04-02 2015-04-01 2,4-thiazolidinedione derivatives in the treatment of central nervous system disorders
EP15713499.0A Active EP3125888B1 (en) 2014-04-02 2015-04-01 2,4-thiazolidinedione derivatives in the treatment of central nervous system disorders

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP15713499.0A Active EP3125888B1 (en) 2014-04-02 2015-04-01 2,4-thiazolidinedione derivatives in the treatment of central nervous system disorders

Country Status (24)

Country Link
US (3) US9782395B2 (en)
EP (2) EP3388064B1 (en)
JP (2) JP6745726B2 (en)
KR (1) KR102418167B1 (en)
CN (1) CN106470991B (en)
AU (1) AU2015239112C1 (en)
CA (1) CA2943373C (en)
CL (1) CL2016002508A1 (en)
CY (2) CY1120454T1 (en)
DK (2) DK3388064T3 (en)
EA (2) EA035866B1 (en)
ES (2) ES2678046T3 (en)
HR (2) HRP20180983T1 (en)
HU (2) HUE039555T2 (en)
IL (1) IL248037B (en)
LT (2) LT3388064T (en)
MX (1) MX2016012687A (en)
NZ (1) NZ725492A (en)
PL (2) PL3125888T3 (en)
PT (2) PT3125888T (en)
SG (1) SG11201608218QA (en)
SI (2) SI3125888T1 (en)
WO (1) WO2015150476A1 (en)
ZA (1) ZA201607286B (en)

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8722710B2 (en) 2007-09-26 2014-05-13 Deuterx, Llc Deuterium-enriched pioglitazone
US20130158077A1 (en) 2011-12-19 2013-06-20 Ares Trading S.A. Pharmaceutical compositions
JP6800013B2 (en) 2013-03-14 2020-12-16 ポクセル・ソシエテ・アノニムPoxel SA Deuterated 2,4-thiazolidinedione and treatment methods
US10188639B2 (en) 2014-01-15 2019-01-29 Deuterx, Llc Methods of treating neurological, metabolic, and other disorders using enantiopure deuterium-enriched pioglitazone
DK3388064T3 (en) * 2014-04-02 2021-04-19 Minoryx Therapeutics S L 2,4-THIAZOLIDINDION DERIVATIVES IN THE CENTRAL NERVOUS DISORDER
ES2867300T3 (en) 2016-12-01 2021-10-20 Minoryx Therapeutics S L 5 - [[4- [2- [5- (1-hydroxyethyl) pyridin-2-yl] ethoxy] phenyl] methyl] -1,3-thiazolidine-2,4-dione for treatment of nonalcoholic fatty liver disease
SI3559010T1 (en) * 2016-12-23 2022-10-28 Minoryx Therapeutics S. L. Process for preparing 5-((4-(2-(5-(1-hydroxyethyl)-2-pyridinyl)ethoxy)phenyl)methyl)-2,4-thiazolidinedione and salts thereof
EP3801516A1 (en) * 2018-06-06 2021-04-14 Minoryx Therapeutics S.L. Method of administering a therapeutically effective amount of 5-[[4-[2-[5-(1-hydroxyethyl)pyridin-2-yl]ethoxy]phenyl]methyl]-1,3-thiazolidine-2,4-dione
CA3102584A1 (en) 2018-06-06 2019-12-12 Minoryx Therapeutics S.L. Use of 5-[[4-[2-[5-acetylpyridin-2-yl]ethoxy]benzyl]-1,3-thiazolidine-2,4-dione and its salts
JP7376934B2 (en) * 2018-06-06 2023-11-09 ミノリックス セラピューティクス エセ.エレ. 5-[[4-[2-[5-(1-hydroxyethyl)pyridin-2-yl]ethoxy]phenyl]methyl]-1,3-thiazolidine-2,4 for use in the treatment of mitochondrial diseases -Diones and their salts
AU2021265345A1 (en) 2020-04-30 2022-12-08 Minoryx Therapeutics S.L. Leriglitazone for treating lung inflammation and interstitial lung disease
US11319313B2 (en) 2020-06-30 2022-05-03 Poxel Sa Crystalline forms of deuterium-enriched pioglitazone
US11767317B1 (en) 2020-06-30 2023-09-26 Poxel Sa Methods of synthesizing enantiopure deuterium-enriched pioglitazone

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5441971A (en) 1991-04-11 1995-08-15 The Upjohn Company Thiazolidinedione derivatives, production and use thereof
CA2106967C (en) * 1991-04-11 2003-12-09 Takashi Sohda Thiazolidinedione derivatives, production and use thereof
AU4104593A (en) * 1992-05-05 1993-11-29 Upjohn Company, The A process for producing pioglitazone metabolite
US5952509A (en) 1996-06-27 1999-09-14 Takeda Chemical Industries, Ltd. Production of benzaldehyde compounds
US6191154B1 (en) 1998-11-27 2001-02-20 Case Western Reserve University Compositions and methods for the treatment of Alzheimer's disease, central nervous system injury, and inflammatory diseases
US6294192B1 (en) 1999-02-26 2001-09-25 Lipocine, Inc. Triglyceride-free compositions and methods for improved delivery of hydrophobic therapeutic agents
US6720001B2 (en) 1999-10-18 2004-04-13 Lipocine, Inc. Emulsion compositions for polyfunctional active ingredients
JP2005514399A (en) 2001-12-21 2005-05-19 スミスクライン ビーチャム コーポレーション Dosage of PPARγ activator
US8993773B2 (en) 2002-07-16 2015-03-31 Cadila Healthcare Limited Process to prepare pioglitazone via several novel intermediates
WO2005016339A1 (en) * 2003-07-24 2005-02-24 Case Western Reserve University Methods for the treatment of parkinson's disease
RU2445957C2 (en) 2006-03-16 2012-03-27 Метаболик Солюшнз Девелопмент Компани Thiazolidinedione analogues
US8722710B2 (en) 2007-09-26 2014-05-13 Deuterx, Llc Deuterium-enriched pioglitazone
EP2512470B1 (en) * 2009-12-15 2016-11-30 Octeta Therapeutics, LLC Ppar-sparing thiazolidinediones and combinations for the treatment of neurodegenerative diseases
US8895536B2 (en) 2010-10-29 2014-11-25 Infirst Healthcare Ltd. Compositions and methods for treating chronic inflammation and inflammatory diseases
WO2013040419A1 (en) * 2011-09-14 2013-03-21 The Trustees Of Columbia University In The City Of New York Compositions and methods for treating hyperprolinemia-associated mental disorders
US8865747B2 (en) * 2012-03-23 2014-10-21 Fundació Institute D'Investigació Biomėdica de Bellvitge (IDIBELL) Pioglitazone for use in the treatment of adrenoleukodystrophy
US20140178456A1 (en) 2012-08-30 2014-06-26 Udaya Sankar Devanaboyina Methods and compositions for treating type 2 diabetes and related conditions
JP6800013B2 (en) * 2013-03-14 2020-12-16 ポクセル・ソシエテ・アノニムPoxel SA Deuterated 2,4-thiazolidinedione and treatment methods
US10188639B2 (en) 2014-01-15 2019-01-29 Deuterx, Llc Methods of treating neurological, metabolic, and other disorders using enantiopure deuterium-enriched pioglitazone
DK3388064T3 (en) * 2014-04-02 2021-04-19 Minoryx Therapeutics S L 2,4-THIAZOLIDINDION DERIVATIVES IN THE CENTRAL NERVOUS DISORDER

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None *

Also Published As

Publication number Publication date
SG11201608218QA (en) 2016-10-28
LT3125888T (en) 2018-07-10
ZA201607286B (en) 2021-07-28
CY1120454T1 (en) 2019-07-10
EP3125888A1 (en) 2017-02-08
PL3388064T3 (en) 2021-07-19
IL248037A0 (en) 2016-11-30
JP2020117539A (en) 2020-08-06
CL2016002508A1 (en) 2017-04-21
BR112016022974A2 (en) 2017-12-26
US10179126B2 (en) 2019-01-15
EP3125888B1 (en) 2018-05-23
CN106470991A (en) 2017-03-01
EA202091120A2 (en) 2020-08-31
AU2015239112A1 (en) 2016-11-10
AU2015239112C1 (en) 2020-08-20
US20180133202A1 (en) 2018-05-17
MX2016012687A (en) 2017-04-27
DK3388064T3 (en) 2021-04-19
DK3125888T3 (en) 2018-06-25
EP3388064A1 (en) 2018-10-17
LT3388064T (en) 2021-05-25
CA2943373A1 (en) 2015-10-08
IL248037B (en) 2020-09-30
HRP20210518T1 (en) 2021-05-14
NZ725492A (en) 2023-06-30
PT3125888T (en) 2018-07-20
ES2872335T3 (en) 2021-11-02
KR20160145639A (en) 2016-12-20
US20190255032A1 (en) 2019-08-22
US20160235729A1 (en) 2016-08-18
JP6745726B2 (en) 2020-08-26
HUE039555T2 (en) 2019-01-28
CY1124088T1 (en) 2022-05-27
EA035866B1 (en) 2020-08-21
US9782395B2 (en) 2017-10-10
PL3125888T3 (en) 2018-09-28
PT3388064T (en) 2021-05-14
CN106470991B (en) 2020-07-31
EA201691997A1 (en) 2017-02-28
AU2015239112B2 (en) 2020-05-07
HRP20180983T1 (en) 2018-08-10
SI3388064T1 (en) 2021-08-31
WO2015150476A1 (en) 2015-10-08
ES2678046T3 (en) 2018-08-08
HUE055351T2 (en) 2022-05-28
EA202091120A3 (en) 2020-12-30
KR102418167B1 (en) 2022-07-08
JP2017511339A (en) 2017-04-20
CA2943373C (en) 2023-01-10
SI3125888T1 (en) 2018-08-31

Similar Documents

Publication Publication Date Title
EP3388064B1 (en) 2,4-thiazolidinedione derivatives in the treatment of central nervous system disorders
US10233178B2 (en) Arylcyclopropylamine based demethylase inhibitors of LSD1 and their medical use
US20110230497A1 (en) Biologically active amides
US20240091210A1 (en) Use of 5-[[4-[2-[5-acetylpyridin-2-yl]ethoxy]benzyl]-1,3-thiazolidine-2,4-dione and its salts
US20070185168A1 (en) Piperidine derivatives
CN115916337A (en) Haloallylamine diamine oxidase inhibitors
EA042526B1 (en) 2,4-THIAZOLIDINEDIONE DERIVATIVES IN THE TREATMENT OF CENTRAL NERVOUS SYSTEM DISORDERS
BR112016022974B1 (en) USE OF A COMPOUND OR MIXTURE OF 2,4-THIAZOLIDINEDIONE DERIVATIVES IN THE TREATMENT OF DISORDERS OF THE CENTRAL NERVOUS SYSTEM
AU2019283649A1 (en) Method of administering a therapeutically effective amount of 5-((4-(2-(5-(1-hydroxyethyl)pyridin-2-yl)ethoxy)phenyl)methyl)-1,3-thiazolidine-2,4-dione
US20240010620A1 (en) Arylazabicyclo[2.1.1]hexylmethanols derivatives and medical uses thereof
US20110039848A1 (en) Five-membered ring compound

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN PUBLISHED

AC Divisional application: reference to earlier application

Ref document number: 3125888

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20190416

RBV Designated contracting states (corrected)

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20191022

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1261676

Country of ref document: HK

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTC Intention to grant announced (deleted)
RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: MINORYX THERAPEUTICS S.L.

INTG Intention to grant announced

Effective date: 20200311

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

INTC Intention to grant announced (deleted)
GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20201002

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AC Divisional application: reference to earlier application

Ref document number: 3125888

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

Ref country code: AT

Ref legal event code: REF

Ref document number: 1369040

Country of ref document: AT

Kind code of ref document: T

Effective date: 20210315

REG Reference to a national code

Ref country code: HR

Ref legal event code: TUEP

Ref document number: P20210518T

Country of ref document: HR

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602015066865

Country of ref document: DE

REG Reference to a national code

Ref country code: FI

Ref legal event code: FGE

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20210412

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20210518T

Country of ref document: HR

Payment date: 20210330

Year of fee payment: 7

REG Reference to a national code

Ref country code: NO

Ref legal event code: T2

Effective date: 20210310

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: RO

Ref legal event code: EPE

REG Reference to a national code

Ref country code: HR

Ref legal event code: T1PR

Ref document number: P20210518

Country of ref document: HR

Ref country code: PT

Ref legal event code: SC4A

Ref document number: 3388064

Country of ref document: PT

Date of ref document: 20210514

Kind code of ref document: T

Free format text: AVAILABILITY OF NATIONAL TRANSLATION

Effective date: 20210507

REG Reference to a national code

Ref country code: GR

Ref legal event code: EP

Ref document number: 20210401086

Country of ref document: GR

Effective date: 20210519

REG Reference to a national code

Ref country code: SK

Ref legal event code: T3

Ref document number: E 36974

Country of ref document: SK

REG Reference to a national code

Ref country code: EE

Ref legal event code: FG4A

Ref document number: E020664

Country of ref document: EE

Effective date: 20210406

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20210310

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20210310

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2872335

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20211102

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20210710

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602015066865

Country of ref document: DE

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20210310

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20210310

26N No opposition filed

Effective date: 20211213

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20210518

Country of ref document: HR

Payment date: 20220331

Year of fee payment: 8

REG Reference to a national code

Ref country code: HU

Ref legal event code: AG4A

Ref document number: E055351

Country of ref document: HU

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20210710

REG Reference to a national code

Ref country code: AT

Ref legal event code: UEP

Ref document number: 1369040

Country of ref document: AT

Kind code of ref document: T

Effective date: 20210310

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20210518

Country of ref document: HR

Payment date: 20230329

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NO

Payment date: 20230327

Year of fee payment: 9

Ref country code: FR

Payment date: 20230320

Year of fee payment: 9

Ref country code: DK

Payment date: 20230328

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: TR

Payment date: 20230331

Year of fee payment: 9

Ref country code: SE

Payment date: 20230315

Year of fee payment: 9

Ref country code: PL

Payment date: 20230327

Year of fee payment: 9

Ref country code: LV

Payment date: 20230320

Year of fee payment: 9

Ref country code: HR

Payment date: 20230329

Year of fee payment: 9

Ref country code: BE

Payment date: 20230322

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: MT

Payment date: 20230323

Year of fee payment: 9

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230525

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IT

Payment date: 20230424

Year of fee payment: 9

Ref country code: ES

Payment date: 20230509

Year of fee payment: 9

Ref country code: DE

Payment date: 20230320

Year of fee payment: 9

Ref country code: CY

Payment date: 20230214

Year of fee payment: 9

Ref country code: CH

Payment date: 20230501

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SI

Payment date: 20230329

Year of fee payment: 9

Ref country code: HU

Payment date: 20230327

Year of fee payment: 9

Ref country code: AT

Payment date: 20230327

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GR

Payment date: 20240326

Year of fee payment: 10

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20210518

Country of ref document: HR

Payment date: 20240320

Year of fee payment: 10

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: LT

Payment date: 20240318

Year of fee payment: 10

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20240319

Year of fee payment: 10

Ref country code: IE

Payment date: 20240326

Year of fee payment: 10

Ref country code: LU

Payment date: 20240319

Year of fee payment: 10

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20210310

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: RO

Payment date: 20240329

Year of fee payment: 10

Ref country code: FI

Payment date: 20240327

Year of fee payment: 10

Ref country code: EE

Payment date: 20240318

Year of fee payment: 10

Ref country code: CZ

Payment date: 20240320

Year of fee payment: 10

Ref country code: BG

Payment date: 20240321

Year of fee payment: 10

Ref country code: PT

Payment date: 20240326

Year of fee payment: 10

Ref country code: GB

Payment date: 20240314

Year of fee payment: 10

Ref country code: SK

Payment date: 20240315

Year of fee payment: 10